US20210205360A1 - Methods and compositions for the treatment of osteopetrosis - Google Patents

Methods and compositions for the treatment of osteopetrosis Download PDF

Info

Publication number
US20210205360A1
US20210205360A1 US17/058,830 US201917058830A US2021205360A1 US 20210205360 A1 US20210205360 A1 US 20210205360A1 US 201917058830 A US201917058830 A US 201917058830A US 2021205360 A1 US2021205360 A1 US 2021205360A1
Authority
US
United States
Prior art keywords
mice
csf1r
cre
bone
cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US17/058,830
Inventor
Frederic GEISSMANN
James T. MULLER
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Memorial Sloan Kettering Cancer Center
Original Assignee
Memorial Sloan Kettering Cancer Center
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Memorial Sloan Kettering Cancer Center filed Critical Memorial Sloan Kettering Cancer Center
Priority to US17/058,830 priority Critical patent/US20210205360A1/en
Publication of US20210205360A1 publication Critical patent/US20210205360A1/en
Assigned to NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT reassignment NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: SLOAN-KETTERING INST CAN RESEARCH
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/15Cells of the myeloid line, e.g. granulocytes, basophils, eosinophils, neutrophils, leucocytes, monocytes, macrophages or mast cells; Myeloid precursor cells; Antigen-presenting cells, e.g. dendritic cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4614Monocytes; Macrophages
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/462Cellular immunotherapy characterized by the effect or the function of the cells
    • A61K39/4622Antigen presenting cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0645Macrophages, e.g. Kuepfer cells in the liver; Monocytes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0654Osteocytes, Osteoblasts, Odontocytes; Bones, Teeth
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/50Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
    • C12N9/64Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue
    • C12N9/6421Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue from mammals
    • C12N9/6472Cysteine endopeptidases (3.4.22)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/31Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/38Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy

Definitions

  • the present technology generally relates to methods and compositions for treating, preventing, or ameliorating osteopetrosis. More specifically, the present technology relates to administering a composition comprising a therapeutically effective amount of engineered monocytic cells or wild-type monocytic cells from a healthy donor to a subject suffering from or at risk for osteopetrosis.
  • the osteopetrosis comprises one or more of stunted growth, skeletal deformity, increased likelihood of bone fracture, anemia, recurrent infections, hepatosplenomegaly, facial paralysis, abnormal cortical bone morphology, abnormal form of vertebral bodies, abnormal temperature regulation, abnormality of the ribs, abnormality of vertebral epiphysis morphology, bone pain, cranial nerve paralysis, craniosynostosis, hearing impairment, and hypocalcemia.
  • the composition is formulated for intravenous administration by injection, infusion, or transfusion.
  • the subject is a mammal.
  • the mammalian subject is a human.
  • the subject is characterized by a cathepsin K deficiency.
  • the osteopetrosis comprises one or more of stunted growth, skeletal deformity, increased likelihood of bone fracture, anemia, recurrent infections, hepatosplenomegaly, facial paralysis, abnormal cortical bone morphology, abnormal form of vertebral bodies, abnormal temperature regulation, abnormality of the ribs, abnormality of vertebral epiphysis morphology, bone pain, cranial nerve paralysis, craniosynostosis, hearing impairment, and hypocalcemia.
  • the composition is formulated for intravenous administration by injection, infusion, or transfusion.
  • the subject is a mammal.
  • the mammalian subject is a human.
  • the subject is characterized by a cathepsin K deficiency.
  • the monocytic cells are obtained from the subject.
  • FIGS. 1A-1I HSC-derived precursors are dispensable for osteoclasts and bone development.
  • FIG. 1A Top: Representative photographs of teeth of control littermates and Csf1 Cre ; Csf1r fl/fl 3-4 week old mice (right) and controls and Csf1r Cre ; Tnfrsf11a fl/fl mice (left). Bottom left: Representative photographs of leg bones from controls and Csf1r Cre ; Tnfrsf11a fl/fl mice, white arrowhead points to lack of blood cells.
  • FIG. 1D Osteoclast counts in femurs from Flt3r Cre ; Tnfrsf11a fl/fl ; Flt3r Cre ; Csf1r fl/fl , Vav Cre ; Csf1r fl/fl mice and littermates of the indicated ages.
  • FIG. 1I Representative confocal microscopy image of frozen sections from mice in ( FIGS. 1G, 1H ).
  • Statistical significance was analysed with GraphPad Prism using unpaired t-tests and two-way ANOVA with Tukey's multiple comparisons test as indicated in the figure legends. Significance was considered at p value (p) *p ⁇ 0.05; **p ⁇ 0.01; ***p ⁇ 0.001; ****p ⁇ 0.0001.
  • FIGS. 3A-3G In vivo dynamics of osteoclasts.
  • 3D Scatter plots represent the Mean Fluorescent Intensity (MFI) of individual TRAP+ MGC for YFP (Y axis) and tdTomato (X axis), and histograms represent the overlayed distribution of the MFI values for YFP and tdTomato in TRAP+ MGC at the indicated time-points.
  • FIG. 3G A model for development and maintenance of osteoclast syncytia. Data are mean ⁇ SD, circles on graphs represent individual mice, n indicates the number of mice per group. Statistics: two-way ANOVA with Tukey's multiple comparisons test *p ⁇ 0.05 **p ⁇ 0.005.
  • FIGS. 4A-G Rescue of Osteopetrosis.
  • FIG. 4G Histograms represent percentages of YFP+ cells among bone marrow precursors and blood leukocytes in the recipient mice (in FIG. 4D ) at the time of analysis. Data are mean ⁇ SD, circles on graphs represent individual mice, n indicates the number of mice per group. Statistical significance was analyzed with GraphPad Prism using ANOVA with Tukey's multiple comparisons test ***p ⁇ 0.001; ****p ⁇ 0.0001.
  • FIGS. 5A-G Csf1r Cre ; Tnfrsf11a fl/fl : Csf1r fl/fl ; Csf1r Cre , and Tnfrsf11a Cre ; Csf1r fl/fl mice are osteopetrotic.
  • FIGS. 5A-B Representative Computed Tomography scans (nanospect CT) of 4 week old Csf1r Cre ; Tnfrsf11a fl/fl mice arrows point skull deformation, and absence of bone marrow cavity in mutant mice.
  • FIG. 5F Representative computed tomography scan reconstructions (nanospect CT) of 4 weeks-old Tnfrsf11a (Koba)Cre ; Csf1r fl/fl mice. Arrows point to skull deformation, but the presence of bone marrow cavity in mutant mice, as in FIGS. 5A , B.
  • FIG. 5G Representative Computed Tomography scans of Csf1 Cre ; Csf1r fl/fl mice. Arrows shows skull deformation, and absence of bone marrow cavity in mutant mice as in A,B. CT scans and photographs are representative of >10 litters.
  • FIGS. 6A-N Bone histology and Flow cytometry analysis of bone marrow phenotypic KSL, LT-HSCs, ST-HSCs, and MPPs in mice of indicated genotypes.
  • FIG. 6A Young Flt3 Cre ; Csf1r fl/fl and Flt3 Cre ; Tnfrsf11a fl/fl mice have normal long bone: H&TRAP staining of bone sections from 4 week old Flt3r Cre ; Tnfrsf11a fl/fl mice showing normal bone structure and bone marrow cavity.
  • FIG. 6A Young Flt3 Cre ; Csf1r fl/fl and Flt3 Cre ; Tnfrsf11a fl/fl mice have normal long bone: H&TRAP staining of bone sections from 4 week old Flt3r Cre ; Tnfrsf11a fl/fl mice showing normal bone structure and bone marrow cavity.
  • FIGS. 6C H&TRAP staining of bone sections from 4 week old Flt3 Cre ; Csf1r fl/fl mice showing normal bone structure and bone marrow cavity.
  • FIGS. 6D , E Phenotypic long-term hematopoietic stem cells (LT-HSCs) are reduced in aged but not old Vav Cre ; Csf1r fl/ ⁇ mice, ( FIGS. 6F , G) LT-HSC are reduced in in young Csf1r ⁇ / ⁇ and to a lesser extent in young Tnfrsf11a (Wask)Cre ; Csf1r fl/ ⁇ Numbers per 2 femur are shown.
  • FIG. 1HSCs Phenotypic long-term hematopoietic stem cells
  • FIG. 6I H&TRAP staining of bone sections from P7 Tnfrsf11a Cre ; Csf1r fl/fl and littermate control showing absence of the bone marrow cavity.
  • FIG. 6J Flow cytometry plot of Fetal liver at E15.5 (B/C representative results of 3 experiments).
  • FIG. 6M Representative micrographs of femur sections from 4 week old Tnfrsf11a Cre ; Csf1r fl/fl and littermate control stained with hematoxylin/TRAP.
  • LT-HSC Lin ⁇ Kit+ Sca-1+; Kit+ Sca1+CD34 ⁇ F1t3 ⁇ .
  • FIGS. 7A-E Bone histomorphometry in old Flt3r Cre ; Tnfrsf11a fl/fl , Flt3 Cre ; Csf1r fl/fl and Vav Cre ; Csf1r fl/fl mice and control littermates.
  • FIG. 7A Representative Micro CT pictures, genotype is indicated.
  • FIGS. 8A-E Colonization of the bone marrow by Csf1r+ and Flt3+ hematopoietic cells.
  • FIG. 8B YFP labeling efficiency in Flt3 Cre ; Rosa26 LSL-YFP mice analyzed by flow cytometry in the indicated cell populations, and by confocal microscopy on frozen bone sections at the indicated time (Right). Insets show YFP expression on individual osteoclasts.
  • FIG. 8C YFP labeling efficiency in Csf1r Cre ; Rosa26 LSL-YFP mice analyzed as in FIG. 8A .
  • Data from FIGS. 8A , B are representative of at least 3 experiments by time point and genotypes. Circles represent individual mice. Genetic lineage tracing of osteoclasts in ossification centers using Csf1r MeriCreMer ; Rosa26 LSL-YFP .
  • FIGS. 9A-D Tnfrs11a (wask)Cre knock-in mice allow deletion of target genes in fetal macrophages, but not definitive HSC and their progeny in blood and tissues, while Vav cre mice allow deletion of target genes in definitive HSC, but not fetal macrophages.
  • FIG. 9A Bar graphs indicate percentages of eYFP expression by flow cytometry in cells from Tnfrsf11a Cre ; Rosa26 LSL-YFP in the indicated cell types, organs, and time points. Data represent 3 independent experiments n numbers indicated on x axis.
  • FIG. 9A Bar graphs indicate percentages of eYFP expression by flow cytometry in cells from Tnfrsf11a Cre ; Rosa26 LSL-YFP in the indicated cell types, organs, and time points. Data represent 3 independent experiments n numbers indicated on x axis.
  • FIG. 9A Bar graphs indicate percentages of eYFP expression by flow cytometry in cells from Tnfr
  • FIG. 9B Lineage tracing in the fetal liver of Vav cre+ ; tdRFP wt/ki mice n numbers indicated on x axis.
  • FL Fetal Liver
  • HSPC hematopoietic stem and progenitor cell
  • LT-HSC Long-term Hematopoietic Stem Cells
  • MPP Multipotent progenitors
  • PMN polymorphonuclear cells
  • mono monocytes
  • T T cells
  • B B cells
  • PEC peritoneal exudate cells.
  • Data are mean ⁇ SD, circles on graphs represent individual mice, n indicates the number of mice per group.
  • FIGS. 10A-F Bone morphometric and dynamic histomorphometry effects of the deletion of Csf1r in P21 Tnfrsf11a Cre ; Csf1 fl/fl mice.
  • FIGS. 11A-C EdU labeling of bone marrow myeloid cells.
  • FIG. 11B Long term kinetics: (1-240 hrs) EdU is injected as in FIG. 11A and % of EdU+ monocytic cells in bone marrow (top) and blood (bottom) is determined by flow cytometry, showing that labeling of ⁇ 50% of monocytic cells is observed for ⁇ 2 days. Labeled cells are not detectable after 3 days. Circles represent determination from individual mice, data for each time point are pooled from 2-3 independent experiments see source data.
  • FIG. 11C Parabiosis between Csf1r Cre ; Rosa26 LSL-YFP and Csf1r Cre ; Rosa26 LSL-tdTomato pairs as described in ( FIG.
  • Scatter plots represent the Mean Fluorescent Intensity (MFI) of individual TRAP + MGC for YFP (Y axis) and tdTomato (X axis), and histograms represent the overlaid distribution of the MFI values for YFP and tdTomato in TRAP + MGC at the indicated time-points. Data are mean ⁇ SD, circles on graphs represent individual mice, n indicates the number of mice per group.
  • MFI Mean Fluorescent Intensity
  • FIGS. 12A-C FACS analysis of monocyte purification, and blood/bone marrow from transferred Csf1r Cre ; Csf1r fl/fl mice.
  • FIG. 12A Representative flow cytometry plots of purified bone marrow monocytes from magnetic bead based enrichment (MACS), percentage of live YFP+ monocytes is indicated.
  • Experiments A-C are representative of 3 independent experiments.
  • FIGS. 14A-F Csf1r deletion in Tnfrsf11a Cre Csf1r F/F mice results in lack of tissue macrophages and osteoclasts at birth, while definitive HSC and their progeny are present in the fetal liver and blood.
  • FIG. 14A Flow cytometry analysis of F4/80+ cells in brain (microglia) and epidermis (Langerhans cells) in E18.5 Tnfrsf11a Cre +; Csf1r F/F embryos and littermate controls.
  • FIG. 14A Flow cytometry analysis of F4/80+ cells in brain (microglia) and epidermis (Langerhans cells) in E18.5 Tnfrsf11a Cre +; Csf1r F/F embryos and littermate controls.
  • FIGS. 15A-D Bone morphometric effects of the deletion of Csf1r in P21 Tnfrsf11a Cre+ ; Csf1r F/ ⁇ or F/F mice.
  • Bone volume/total volume (BV/TV, FIG. 15A ), Bone length FIG. 15B , connectivity density (Conn density, FIG. 15C ), and Trabecular Number (Tb.N., FIG. 15D ) was analyzed by microCT in 21 day/old mice of indicated genotype.
  • Controls included Tnfrsf11a(wask) Cre+ ; Csf1r F/+ or Tnfrsf11a (koba)Cre ⁇ ; Csf1r F/F and Tnfrsf11a (koba)Cre+ ; Csf1r F/+ types. Circles represent 1 mouse with 3-7 mice per group. For statistical analysis unpaired two-tailed t-test. *p ⁇ 0.05, **p ⁇ 0.005, ***p ⁇ 0.0005, ****p ⁇ 0.0001.
  • the term “about” and the use of ranges in general, whether or not qualified by the term about, means that the number comprehended is not limited to the exact number set forth herein, and is intended to refer to ranges substantially within the quoted range while not departing from the scope of the invention.
  • “about” will be understood by persons of ordinary skill in the art and will vary to some extent on the context in which it is used. If there are uses of the term which are not clear to persons of ordinary skill in the art given the context in which it is used, “about” will mean up to plus or minus 10% of the particular term.
  • a “chimeric nucleic acid” comprises a coding sequence or fragment thereof linked to a nucleotide sequence that is different from the nucleotide sequence with which it is associated in cells in which the coding sequence occurs naturally.
  • engineered is used herein to refer to a cell or organism that has been manipulated to be genetically altered, modified, or changed, e.g., by disruption of the genome.
  • engineered monocytic cell refers to a monocytic cell that has been manipulated to be genetically altered, modified, or changed.
  • Heterologous nucleic acid refers to a nucleic acid, DNA, or RNA, which has been introduced into a cell (or the cell's ancestor), and which is not a copy of a sequence naturally found in the cell into which it is introduced.
  • Such heterologous nucleic acid may comprise segments that are a copy of a sequence that is naturally found in the cell into which it has been introduced, or fragments thereof.
  • prevention refers to, in a statistical sample, reduction in the occurrence or recurrence of the disorder or condition in treated subjects/samples relative to an untreated controls, or refers delays the onset of one or more symptoms of the disorder or condition relative to the untreated controls.
  • compositions and methods for the treatment of osteopetrosis identify: (i) the developmental origin of osteoclasts, the cells that shape bone architecture; (ii) a mechanism that controls the maintenance of osteoclast function after birth; and (iii) a novel strategy to treat osteopetrosis and to modulate osteoclast activity in vivo.
  • the data described herein demonstrate that parabiosis or transfusion of monocytic cells results in long-term gene transfer in osteoclasts in the absence of HSC chimerism and can rescue an adult-onset osteopetrotic phenotype caused by cathepsin-K deficiency. Transfusion of monocytic cells is also sufficient to rescue bone development in early-onset autosomal recessive osteopetrosis in newborn mice.
  • Osteopetroses are a heterogeneous group of genetic disorders characterized by increased bone density due to impaired bone resorption by osteoclasts. The increased bone density places the affected individual at an increased risk for bone fracture. Normally, bone growth is a balance between osteoblasts (cells that create bone tissue) and osteoclasts (cells that destroy bone tissue). Individuals with osteopetrosis have a deficiency of osteoclasts, resulting in too little bone resorption and too much bone creation. The types of osteopetrosis are distinguished based on their pattern of inheritance: autosomal dominant, autosomal recessive, or X-linked.
  • the signs and symptoms of osteopetrosis may vary depending on the type of the disease, and mild forms of osteopetrosis may be asymptomatic.
  • the typical signs and symptoms of osteopetrosis include: stunted growth, skeletal deformity, increased likelihood of bone fracture, anemia, recurrent infections, hepatosplenomegaly, facial paralysis, abnormal cortical bone morphology, abnormal form of vertebral bodies, abnormal temperature regulation, abnormality of the ribs, abnormality of vertebral epiphysis morphology, bone pain, cranial nerve paralysis, craniosynostosis, hearing impairment, and hypocalcemia.
  • ADO autosomal dominant osteopetrosis
  • Albers-Schonberg disease The signs and symptoms of autosomal dominant osteopetrosis (ADO; also known as Albers-Schonberg disease), include multiple bone fractures, scoliosis, arthritis in the hips, and/or osteomyelitis.
  • Autsomal recessive osteopetrosis (ARO) is often characterized by one or more of a high risk of bone fracture resulting from minor bumps or falls, pinched nerves in the head and face, impaired bone marrow function, slow growth, short stature, dental abnormalities, hepatosplenomegaly, intellectual disability, and epilepsy.
  • IAO intermediate autosomal osteopetrosis
  • a form of osteopetrosis that can have either an autosomal dominant or recessive pattern or inheritance may be characterized by one or more of a high risk of bone fracture and anemia, calcifications in the brain, intellectual disability, and renal tubular acidosis.
  • Individuals with the X-linked pattern of inheritance may be characterized by one or more of lymphedema, anhidrotic ectodermal dysplasia, and immunodeficiency.
  • a method for treating or preventing osteopetrosis in a subject in need thereof, comprising administering a composition comprising a therapeutically effective amount of monocytic cells from a healthy donor to the subject comprising administering a composition comprising a therapeutically effective amount of monocytic cells from a healthy donor to the subject.
  • the subject is characterized by decreased expression of one or more genes implicated in or potentially underlying the development of osteopetrosis, such as CA2, CLCN7, CTSK, CSF1R, IKBKG, ITGB3, OSTM1, PLEKHM1, TCIRG1, TNFRSF11A, and TNFSF11.
  • the subject is characterized by a cathepsin K deficiency.
  • a method for treating or preventing osteopetrosis in a subject in need thereof comprising administering to the subject a composition comprising a therapeutically effective amount of monocytic cells engineered to express one or more genes implicated in or potentially underlying the development of osteopetrosis, such as CA2, CLCN7, CTSK, CSF1R, IKBKG, ITGB3, OSTM1, PLEKHM1, TCIRG1, TNFRSF11A, and TNFSF11 is provided.
  • monocytic cells used for treatment are genetically modified to correct a genetic abnormality or to improve or changed cellular functioning according to known genetic engineering protocols.
  • a method of treating or preventing osteopetrosis in a subject comprises: (a) obtaining a sample of monocytic cells from the subject; (b) genetically correcting one or more mutations in the monocytic cells, (c) culturing the monocytic cells; and (d) providing the corrected monocytic cells to the subject.
  • a method for treating or preventing osteopetrosis comprises: (a) obtaining a sample of monocytic cells from the subject; (b) genetically engineering the cells to express a nucleotide sequence coding for the wild type allele of any one or more of CA2, CLCN7, CTSK, CSF1R, IKBKG, ITGB3, OSTM1, PLEKHM1, TCIRG1, TNFRSF11A, and TNFSF11 with a suitable transducing vector, such as a lentiviral vector; (c) culturing the engineered monocytic cells under conditions sufficient to express the nucleotide sequence; (d) removing the viral particles from the engineered monocytic cells; and (e) providing the engineered monocytic cells to the subject.
  • a suitable transducing vector such as a lentiviral vector
  • the transducing vector encoding the one or more of CA2, CLCN7, CTSK, CSF1R, IKBKG, ITGB3, OSTM1, PLEKHM1, TCIRG1, TNFRSF11A, and TNFSF11 is a mammalian expression vector.
  • the mammalian expression vector is a lentiviral vector or transposon vector.
  • Subjects suffering from osteopetrosis can be identified by any or a combination of diagnostic or prognostic assays known in the art.
  • typical symptoms of osteopetrosis include, but are not limited to, stunted growth, skeletal deformity, increased likelihood of bone fracture, anemia, recurrent infections, hepatosplenomegaly, facial paralysis, abnormal cortical bone morphology, abnormal form of vertebral bodies, abnormal temperature regulation, abnormality of the ribs, abnormality of vertebral epiphysis morphology, bone pain, cranial nerve paralysis, craniosynostosis, hearing impairment, and hypocalcemia.
  • a composition comprising a therapeutically effective amount of monocytic cells from a health donor and/or monocytic cells engineered to express one or more genes selected from CA2, CLCN7, CTSK, CSF1R, IKBKG, ITGB3, OSTM1, PLEKHM1, TCIRG1, TNFRSF11A, and TNFSF11 is administered to the subject.
  • the composition is administered according to any acceptable transfusion regimen.
  • the composition is administered one, two, three, four, or five times per day.
  • the composition is administered more than five times per day.
  • the composition is administered every day, every other day, every third day, every fourth day, every fifth day, or every sixth day.
  • the composition is administered weekly, bi-weekly, tri-weekly, or monthly. In some embodiments, the composition is administered for a period of one, two, three, four, or five weeks. In some embodiments, the composition is administered for six weeks or more. In some embodiments, the composition is administered for twelve weeks or more. In some embodiments, the composition is administered for a period of less than one year. In some embodiments, the composition is administered for a period of more than one year or until a desired therapeutic outcome is observed in the subject.
  • treatment of subjects diagnosed with or suspected of having osteopetrosis with one or more compositions of the present technology ameliorates or eliminates one or more of the following symptoms of osteopetrosis: stunted growth, skeletal deformity, increased likelihood of bone fracture, anemia, recurrent infections, hepatosplenomegaly, facial paralysis, abnormal cortical bone morphology, abnormal form of vertebral bodies, abnormal temperature regulation, abnormality of the ribs, abnormality of vertebral epiphysis morphology, bone pain, cranial nerve paralysis, craniosynostosis, hearing impairment, and hypocalcemia.
  • the present technology provides a method for preventing or delaying the onset of osteopetrosis or one or more symptoms of osteopetrosis in a subject at risk of having or developing osteopetrosis.
  • compositions of the present technology are administered to a subject susceptible to, or otherwise at risk of for osteopetrosis in an amount sufficient to eliminate or reduce the risk, or delay the onset of the disease, including biochemical, histologic and/or behavioral symptoms of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease.
  • Administration of a prophylactic compositions can occur prior to the manifestation of symptoms characteristic of the disease or disorder, such that the disease or disorder is prevented or, alternatively, delayed in its progression.
  • Csf1r iCre , Csf1r MeriCreMer , Csf1r flox were kindly provided by Dr Jeffrey Pollard
  • Flt3 Cre were kindly provided by Dr Thomas Boehm
  • Myb +/ ⁇ mice were kindly provided by Dr John Frampton
  • Vav Cre were kindly provided from Dr Thomas Graf.
  • Tnfrsf11a flox mice were kindly provided by J M Penninger
  • Tnfrsf11a koba/Cre mice were kindly provided by Dr Yasuhiro Kobayashi
  • Ctsk tm1(cre)Ska mice were kindly provided by Dr.
  • Rosa.26-CreER T2 (R26-CreER T2 ) were kindly provided from Drs. Pierre Chambon and Anton Berns. Rosa26 LSL-YEP (stock number: 006148) and Rosa26 LSL-tdTomato (stock number: 007908) reporter mice were purchased from The Jackson Laboratory.
  • mice were bred and kept under specific pathogen conditions in separated ventilated cages in the animal facility of MSKCC and the Medical Theoretical Center of the TU Dresden. All experiments with osteopetrotic mice that lack teeth were performed with mice maximal 4 weeks of age that were kept with the lactating mother or provided with DietGel 76A (Clear H 2 O, 72-07-5022) to avoid secondary effects from malnutrition. Experiments were performed in adherence to the Institutional Review Board (IACUC 15-04-006) from MSKCC and Austindiretation Dresden and were in compliance with relevant ethical regulations. Mice greater than 7 days old were sacrificed by cervical dislocation (TU Dresden), CO2 asphyxiation or anesthesia (MSKCC). To harvest embryos, pregnant females were sacrificed and embryos were collected by postmortem cesarean from the uterus and exsanguinated through decapitation in cold PBS (Fisher, 14190).
  • Genotyping PCR genotyping was performed according to protocols described previously and indicated in Table 1. The investigators were not blinded to allocation during experiments and outcome assessment.
  • mice were crossed at night, the following day a positive vaginal plug was considered as 0.5 days post-coitum (dpc), as previously described. Embryos were harvested as indicated below. For postnatal time point, pregnant females were monitored for the date of delivery, caesarean sections were carried out at term and neonates were fostered using lactating CD-1 females.
  • Csf1 iCre mice were crossed to male Rosa26 LSL-YFP/YFP or Rosa26 LSL-tdTomato/tdTomato mice.
  • Csf1r iCre ; Rosa26 LSL-YFP and Csf1r iCre ; Rosa26 LSL-tdTomato females were used for parabiosis.
  • Parabionts were kept on Sulfamethoxazole/Trimethoprim (Sulfatrim) diet for up to 8 weeks. Ex-parabionts were separated after 4 weeks for cellular complementation and 6 weeks for rescue of CathepsinK activity.
  • anesthetic agent bupivacaine (Marcaine 0.25-0.5% solution) was applied locally. Surgery was performed by a longitudinal skin incision on the lateral side of mice, approximately 0.5 cm above the elbow to 0.5 cm below the knee joint. Mice were laid side-by-side in close contact and the ligaments of the two knees and elbows were sutured together using monofilament non-absorbable suture. Then, the skin incisions was closed by apposing and clipping skin to skin of the pair with 9 mm wound clips.
  • mice Post-operative procedure: immediately after surgery mice were injected subcutaneously with 2 mg/kg of meloxicam and 0.5 mg/kg of buprenorphine and for a maximum of 48 hours postoperatively. Mice are provided with Sulfatrim and Hydrogel (Clear H 2 O, 70-01-5022) in medicups. Wound clips were removed 14 days after surgery under Isofluorane anesthesia.
  • Bones were prepared for histology on frozen sections as detailed below, and stained with antibodies to detect fluorescent proteins: anti-GFP biotin, anti-RFP (Abcam), fluorescent TRAP staining and TO-PRO-3 as a nuclear stain. Histological sections of 15 ⁇ m thickness were scanned by confocal microscopy at 1.5 ⁇ m Z stacks. Sections were quantified for the number of TRAP + multinuclear cells (more than 3 nuclei per cell) and their YFP and Tomato expression using Imaris in 3D view and individual Z stacks.
  • EdU pulse labeling 12 week old C57BL/6N mice (Charles River) were injected intraperitoneally with 25 ⁇ g/g of a 2.5 mg/mL solution of EdU prepared extemporaneously (Fisher C10420, Click-iT EdU Alexa Fluor 488 Flow Cytometry Assay Kit). Blood, bone marrow, and bone samples were collected at 60 hrs and 72 hrs post injection and bone were prepared for histology of frozen sections as indicated above for adult mice.
  • Bone marrow monocytes were isolated from 12-16 weeks old donor. In fate-mapping experiments 1 ⁇ 10 6 total cells from Csf1r iCre ; Rosa26 LSL-tdTomato mice were transferred at day 0, day 3 and day 6 by retro-orbital injections into recipient Csf1r iCre ; Rosa26 LSL-YFP age and gender matched mice. Mice were sacrificed at day 11 and day 60 after the first transfer.
  • Bone samples were fixed in 4% formaldehyde (Fisher, 28908) in PBS for 1 day (embryo) or 3 days (post-natal mice) at 4° C. then washed 3 times with PBS and decalcified (for mice older than P7) in a 14% EDTA pH7.1 solution at 4° C. for 5 days to 15 days, washed 3 times with PBS and dehydrated in 70% ethanol for 1 day and processed for paraffin sections.
  • Longitudinal sections of femurs were cut at 5 ⁇ m thickness using a Leica RM2265 paraffin microtome then place in Superfrost microscope slides let dry for 48 hours and heated in a dry incubator at 65° C. for 1 hour, dewaxed and stained for TRAP and hematoxylin.
  • TRAP Tartrate Resistant Acid Phosphatase staining protocol.
  • slides were placed in coplin jars and incubated in a 1% (v/v) Naphtol-Ether substrate solution in basic stock solution for 1 hr at 37° C. followed by incubation in a solution containing 2% (v/v) sodium nitrate solution and 2% (v/v) basic fuchsin solution in basic stock solution for 20 mins at 37° C.
  • Napthol-Ether substrate solution 2% (w/v) Napthol AS-BI. Phosphate (Sigma, 70482) in 2-Ethoxyethanol (Sigma, 256374).
  • Sodium nitrate solution 4% (w/v) Sodium nitrate (Sigma, 237213) in water.
  • Basic fuchsin solution 5% (w/v) basic fuchsin dye (Sigma, 857343) in 2N HCL (Fisher, A144-500).
  • bones were embedded in methacrylate and cut into 7 ⁇ m sections to assess the fluorescent calcein labels. Unstained sections were analyzed using fluorescence microscopy to determine the mineralized surface/bone surface (MS/BS), the mineral apposition rate (MAR), and the bone formation rate/bone surface (BFR/BS). To determine numbers of osteoclasts, bones were decalcified for one week using Osteosoft (Merck), dehydrated, and embedded into paraffin. Tartrate-resistant acid phosphatase staining was used to assess the osteoclast surface per bone surface (Oc.SBS) and number of osteoclasts per bone surface (N.OcBS). Bone sections were analyzed using the Osteomeasure software (Osteometrics, USA) following international standards.
  • Osteomeasure software Osteometrics, USA
  • Sections were washed 2 times with PBS for 5 minutes. Sections stained with anti-GFP biotin antibody were first incubated with Biotin/Streptavidin blocking kit (Vector laboratories, SP2002). Streptavidin blocking solution is prepared by adding 4 drops of streptavidin solution to 1 mL of PBS/0.25% BSA, samples were incubated for 15 min, then washed once with PBS for 5 min. Biotin blocking solution is prepared by adding 4 drops of biotin solution to 1 mL of PBS/0.25% BSA, samples were incubated for 15 min then washed once with PBS for 5 min. Primary and secondary antibodies used are listed in Table 2. Sections were also stained with fluorescent TRAP and nuclear stain.
  • mice were sacrificed and bones placed in 70% ethanol until scanning. Bone microarchitecture was analyzed using the vivaCT40 (Scanco Medical, Switzerland). Entire femora or humeri were imaged at a resolution of 10.5 ⁇ m (1 slice) with an X-ray energy of 70 kVp, 114 mA, and an integration time of 200 ms. The machine was routinely calibrated using hydroxyapatite phantoms for density and geometry. Trabecular bone in femora or humeri from old mice was assessed in the metaphysis 20 slices below the growth plate using 150 slices. The trabecular region within the cortical bones (P21 mice) was determined in the femoral midshaft (100 slices up, 100 slices down). Pre-defined scripts from Scanco were used for the evaluation.
  • Yolk sac from E10.5 embryos was digested for 60 min at 37° C. in PBS containing 5% FCS, Collagenase Type 4 (Worthington, final 4.2 U/ml) and DNAseI (Sigma-Aldrich, final 100 ⁇ g/ml). The digestion reaction was stopped by incubation with 12.5 mM EDTA. Fetal liver was gently dissociated between the frosted ends of glass slides, and was then digested for 30 minutes using the same digestion enzyme mix as yolk sac.
  • Blood was collected from anesthetized mice by retro-orbital venous sinus bleeding or cardiac puncture using a 1 mL syringe and a 26G needle rinsed with 100 mM EDTA (sigma E4884).
  • the collected blood was lysed with 3 mL of red blood cell lysis buffer (155 mM NH 4 Cl sigma A9434, 10 mM NaHCO 3 sigma S5761 and 0.1 mM EDTA sigma E4884) for 5 mins, washed with 10 mL of FACS buffer (PBS, 0.5% BSA and 2 mM EDTA) and centrifuged at 320 g for 7 minutes at 4° C.
  • red blood cell lysis buffer 155 mM NH 4 Cl sigma A9434, 10 mM NaHCO 3 sigma S5761 and 0.1 mM EDTA sigma E4884
  • FACS buffer PBS, 0.5% BSA and 2 mM EDTA
  • Bone marrow cells were harvested by crushing or flushing femurs and tibias using a syringe and a 26G needle with 10 mL of PBS/5% FCS or RPMI supplemented with 10% FBS. Bone marrow was dissociated by gently pipetting up and down with a 1 mL or 10 mL pipette. Spleens were gently dispersed between frosted slides and digested for 30 min at 37° C.
  • the cell pellets were resuspended in FACS buffer containing purified anti-mouse CD16/32 antibody (1:100, Biolegend 101301), 5% normal mouse (Fisher, 015-000-120), 5% normal rat (Fisher 012-000-120) and 5% normal rabbit serum (Fisher, 011-000-120) and incubated for 10 minutes on ice or directly stained using blocking antibodies in the staining mixtures.
  • Samples were immunostained with fluorochrome-conjugated antibodies for 30 minutes on ice, and analyzed by flow cytometry using an LSRFortessa or an LSR II (BD-Bioscience). Full list of antibodies for flow cytometry is provided in Table 3. Each sample was stained with Hoechst (ThermoFisher Scientific, Hoechst 33258, 1 ⁇ g/ml) or DAPI (Applichem, 1 ⁇ g/25 ml) moments prior to flow cytometry acquisition.
  • Hoechst ThermoFisher Scientific
  • CD3 145-2C11 bio eBioscience 1/1000 (BM, Y, FL) CD3 145-2C11 BV711 BD 1/100 (BM, SP, LV, K, BR) Pharmingen 1/400
  • E CD3 145-2C11 APC-Cy7 BD 1/100 (BL) Pharmingen CD3 145-2C11 PE Biolegend 1/100 (BM) CD3 17A2 eF450 eBioscience 1/200
  • CD11b M1/70 bio eBioscience 1/1000 (BM, Y, FL) CD11b M1/70 PE-Cy7 eBioscience 1/200
  • BM, SP, LV, K, BR, LN E
  • BM CD19 1D3 bio eBioscience 1/500
  • BM, Y, FL CD19 1D3 BV711 BD 1/100 (BM, SP, LV, K, K,
  • mice presented with an osteopetrotic phenotype similar to Csf1 (op/op), Csf1r, and Tnfrsf11a mutants and characterized in young mice by lack of teeth eruption, skull and skeletal deformities with shortness of long bones, increased bone density, and lack of osteoclasts and hematopoietic cells ( FIG. 1A ; FIG. 5B ).
  • FIG. 1B hematopoietic cell numbers in long bones at 4 week of age
  • FIG. 1C hematopoietic cell numbers in long bones at 4 week of age
  • Csf1r Cre and Flt3 Cre Fetal expression of Csf1r Cre and Flt3 Cre was analyzed using a fluorescent reporter in Csf1r Cre ; Rosa26 LSL-YFP and Flt3 Cre ; Rosa26 LSL-YFP mice ( FIG. 1 G-I).
  • TRAP + multinucleated cells that appear at embryonic day (E)15 in ossification centers are labeled with YFP in Csf1r Cre ; Rosa26 LSL-YFP mice and osteoclasts remain YFP positive throughout life ( FIG. 1 G,I; FIG.
  • TRAP + multinucleated cells develop in ⁇ E15 ossification centers from Myb-deficient embryo, which lack HSC but still support the development of EMP-derived macrophages ( FIG. 2A , B).
  • TRAP + multinucleated cells are labeled with YFP in Csf1r Mer-iCre-Mer ; Rosa26 LSL-YFP mice pulsed at E8.5 with a single dose of hydroxytamoxifen (4-OHT), which labels EMP but not HSC ( FIG. 2C , D; FIG. 8A-E ).
  • Tnfrsf11a is expressed by osteoclasts, but its expression is also a hallmark of EMP-derived pMacs that colonize the developing embryo.
  • Cre-mediated expression of a Rosa26 LSL-YFP fluorescent reporter is achieved with high efficiency in fetal macrophages but with low efficiency or not at all in HSC and their progeny in blood and tissues ( FIG. 9A-D ). It was therefore hypothesized that conditional deletion of Csf1r in Tnfrsf11a Cre would recapitulate macrophage deficiency observed in Csf1r-deficient mice, while leaving the HSC lineage unaffected.
  • Tnfrsf11a Cre To test osteoclast and bone development in this model, two independent lines of Tnfrsf11a Cre ; Csf1r fl/fl mice were generated ( FIG. 2E-J ). Tnfrsf11a Cre ; Csf1r fl/fl lack tissue macrophages such as brain microglia and epidermal Langerhans cells at birth while development of HSCs and blood cells was preserved ( FIG. 6A-N ). They present with a severe osteopetrotic phenotype including the lack of tooth eruption ( FIG. 2E ), misshaped skulls and short long bones ( FIG. 2F-H ; FIG. 5A-G ) with few osteoclasts ( FIG.
  • FIG. 3A To probe the mechanisms that underlie the contribution of HSC-derived blood cells to osteoclast maintenance as well as the lifespan and dynamics of osteoclasts in vivo, time-course parabiosis experiments were performed ( FIG. 3A ). After 4 to 8 weeks of shared blood circulation between Csf1r Cre ; Rosa26 LSL-YFP and Csf1r Cre ; Rosa26 LSL-tdTomato parabionts, all osteoclasts, defined as TRAP + multinucleated cells lining the bone surface, expressed both YFP and tdTomato within the same cells ( FIG. 3A , B). No other cell was found to co-express YFP and tdTomato in bones ( FIG. 3B ).
  • FIG. 3F suggesting acquisition by osteoclasts of single post-mitotic nuclei at a time.
  • the proportion of osteoclasts acquiring a new nucleus can be estimated ⁇ 0.5 to 2% per day in this model, compatible with individual nuclei being replaced every ⁇ 2 months.
  • FIG. 3G the studies above suggested a model ( FIG. 3G ) where: osteoclasts that control skeletal development develop in ossification centers from EPM, while their post-natal maintenance is mediated via the serial acquisition by long-lived syncytia of new nuclei from HSC-derived blood leukocytes, rather than by de novo renewal by lateral fusion or proliferation of osteoclast precursors.
  • a prediction from this model is that osteopetrosis due to a recessive mutation affecting osteoclasts function may be rescued or prevented through parabiosis with a wild-type partner.
  • Parabiosis experiments between 4 week-old cathepsin K deficient mice, which develop an adult-onset form of osteopetrosis known as pycnodysostosis, and cathepsin K +/ ⁇ or cathepsin K ⁇ / ⁇ littermates and between wild type mice as control showed a reduction of bone volume in 10 weeks old cathepsin K ⁇ / ⁇ mice paired with cathepsin K +/ ⁇ littermates ( FIG. 4A ), suggesting that circulating blood cells carrying a wild type cathepsin K allele are sufficient to reduce bone density.
  • FIG. 4D histology of femurs indicated the presence of numerous YFP+ TRAP+ osteoclasts lining the bone ( FIG. 4E , F). Histology and flow cytometry analyses showed the lack of YFP+ circulating blood cells or bone marrow progenitors ( FIG. 4E , G), indicating absence of HSC engraftment.
  • osteoclasts originating from EMP are essential for normal bone development. Moreover, it is shown that osteoclasts are long-lived in adult and their function is maintained by iterative fusion of individual HSC-derived circulating cells with existing syncytia. In the absence or deficiency of EMP-derived osteoclasts, however, their timely replacement by transfusion with monocytic cells can rescue bone development in early-onset osteopetrotic mice in the absence of bone marrow transplantation. This could contribute to the treatment of early onset osteopetrosis, since the current treatment by bone marrow transplantation.
  • Example 3 Ex Vivo Gene Therapy for the Treatment of a Form of Osteopetrosis: Pycnodysostosis
  • This example demonstrates the prophetic use of monocytic cells obtained from a subject and engineered to express a gene, such as CTSK, for the treatment of osteopetrosis in the subject.
  • Pycnodysostosis is an autosomal recessive form of osteopetrosis, due to loss of function mutations in the cathepsin K gene (CTSK), compatible with life, and characterized by short stature, deformity of the skull maxilla and phalanges, increased density of the bones, osteosclerosis, and fragility of bone. There is no treatment for this disease as the relatively comparably milder symptoms do not qualify for bone marrow transplantation.
  • Subjects suspected of having or diagnosed as having cathepsin K deficiency will be selected for a gene therapy trial and will undergo GMP grade collection of peripheral blood monocytes by apheresis and/or elutriation. Purified monocytes will then be transduced with a lentivirus designed to express a cDNA coding for the wild type allele of CTSK under a strong promoter (e.g., CMV). Transduced monocytes will then be washed to remove contaminating viral particles and rested for 12 hours followed by reinfusion to the patient. Assuming ⁇ 10 9 monocytes can be collected, the number of cells transferred per kilogram of body weight would be >10′. Following the auto-transfusion, the subject will be monitored at regular intervals for clinical and radiological signs of pycnodysostosis in order to determine the efficiency of the procedure and the frequency with which it should be repeated.
  • a strong promoter e.g., CMV
  • Example 4 Ex Vivo Gene Therapy for the Treatment of a Form of Osteopetrosis: Severe Infantile Autosomal Recessive Osteopetrosis
  • This example demonstrates the prophetic use of monocytic cells obtained from a subject and engineered to express a gene, such as TCIRG1, TNFRSF11a, CA2, CLCN7, OSTM1, and/or PLEKHM1, for the treatment of osteopetrosis in the subject.
  • a gene such as TCIRG1, TNFRSF11a, CA2, CLCN7, OSTM1, and/or PLEKHM1, for the treatment of osteopetrosis in the subject.

Abstract

Disclosed herein are methods and compositions for treating, preventing, or ameliorating osteopetrosis. In some embodiments, the present technology relates to administering a composition comprising a therapeutically effective amount of engineered monocytic cells or wild-type monocytic cells from a healthy donor to a subject suffering from or at risk for osteopetrosis.

Description

    CROSS-REFERENCE TO RELATED APPLICATION
  • The present application claims priority to U.S. Provisional Patent Application No. 62/679,553, filed on Jun. 1, 2018, the contents of which are hereby incorporated by reference in their entirety.
  • STATEMENT OF FEDERALLY FUNDED RESEARCH
  • This invention was made with government support under CA008748, AI130345 and HL138090 awarded by the National Institutes of Health. The government has certain rights in the invention.
  • TECHNICAL FIELD
  • The present technology generally relates to methods and compositions for treating, preventing, or ameliorating osteopetrosis. More specifically, the present technology relates to administering a composition comprising a therapeutically effective amount of engineered monocytic cells or wild-type monocytic cells from a healthy donor to a subject suffering from or at risk for osteopetrosis.
  • BACKGROUND
  • The following description is provided to assist the understanding of the reader. None of the information provided or references cited is admitted to be prior art.
  • Osteoclasts are multinucleated giant cells that resorb bone, ensuring development and continuous remodeling of the skeleton and the bone marrow hematopoietic niche. Defective osteoclast activity leads to osteopetrosis and bone marrow failure, while excess activity can contribute to bone loss and osteoporosis. Osteopetrosis can be partially treated by bone marrow transplantation in human and mice, in accordance with osteoclasts hematopoietic origin, and studies suggesting that they develop by fusion of hematopoietic stem cell (HSC)-derived monocytic precursors in the presence of CSF1 and RANK-Ligand. However, the developmental origin and lifespan of osteoclasts, and the mechanisms that ensure maintenance of osteoclast function throughout life in vivo remain largely unexplored. Moreover, there is a need to develop alternative therapeutic approaches to the treatment of osteopetrosis as the current treatment by bone marrow transplantation is plagued by an approximate 48% overall survival at 6 years.
  • SUMMARY
  • In one aspect, the present disclosure provides a method for treating or preventing osteopetrosis in a subject in need thereof, the method comprising administering a composition comprising a therapeutically effective amount of monocytic cells from a healthy donor to the subject. In some embodiments, the subject is characterized by decreased expression of one or more of CA2, CLCN7, CTSK, CSF1R, IKBKG, ITGB3, OSTM1, PLEKHM1, TCIRG1, TNFRSF11A, and TNFSF11, as compared to the monocytic cells of the donor. In some embodiments, the osteopetrosis comprises one or more of stunted growth, skeletal deformity, increased likelihood of bone fracture, anemia, recurrent infections, hepatosplenomegaly, facial paralysis, abnormal cortical bone morphology, abnormal form of vertebral bodies, abnormal temperature regulation, abnormality of the ribs, abnormality of vertebral epiphysis morphology, bone pain, cranial nerve paralysis, craniosynostosis, hearing impairment, and hypocalcemia. In some embodiments, the composition is formulated for intravenous administration by injection, infusion, or transfusion. In some embodiments, the subject is a mammal. In some embodiments, the mammalian subject is a human. In some embodiments, the subject is characterized by a cathepsin K deficiency.
  • In one aspect, the present disclosure provides a method for treating or preventing osteopetrosis in a subject in need thereof, the method comprising administering to the subject a composition comprising a therapeutically effective amount of monocytic cells engineered to express one or more genes selected from CA2, CLCN7, CTSK, CSF1R, IKBKG, ITGB3, OSTM1, PLEKHM1, TCIRG1, TNFRSF11A, and TNFSF11. In some embodiments, the osteopetrosis comprises one or more of stunted growth, skeletal deformity, increased likelihood of bone fracture, anemia, recurrent infections, hepatosplenomegaly, facial paralysis, abnormal cortical bone morphology, abnormal form of vertebral bodies, abnormal temperature regulation, abnormality of the ribs, abnormality of vertebral epiphysis morphology, bone pain, cranial nerve paralysis, craniosynostosis, hearing impairment, and hypocalcemia. In some embodiments, the composition is formulated for intravenous administration by injection, infusion, or transfusion. In some embodiments, the subject is a mammal. In some embodiments, the mammalian subject is a human. In some embodiments, the subject is characterized by a cathepsin K deficiency. In some embodiments, the monocytic cells are obtained from the subject.
  • In one aspect, the present disclosure provides a donor monocytic cell line engineered to express one or more genes selected from CA2, CLCN7, CTSK, CSF1R, IKBKG, ITGB3, OSTM1, PLEKHM1, TCIRG1, TNFRSF11A, and TNFSF11, wherein the one or more genes is operably linked to a heterologous nucleic acid to form a chimeric nucleic acid construct. In some embodiments, the heterologous nucleic acid encodes a selectable marker. In some embodiments, the selectable marker is a bioluminescent protein, a fluorescent protein, a chemiluminescent protein, a xanthine-guanine phosphoribosyl transferase gene (gpt), or any combination thereof. In some embodiments, the heterologous nucleic acid encodes one or more control sequences suitable for directing expression of the one or more genes in a monocytic cell. In some embodiments, the one or more control sequences comprises a promoter. In some embodiments, the donor cells comprise a vector encoding one or more genes selected from CA2, CLCN7, CTSK, CSF1R, IKBKG, ITGB3, OSTM1, PLEKHM1, TCIRG1, TNFRSF11A, and TNFSF11. In some embodiments, the vector is a mammalian expression vector, a lentiviral vector, or transposon vector.
  • The technology described and claimed herein has many attributes and embodiments including, but not limited to, those set forth or described or referenced in this brief summary. It is not intended to be all-inclusive and the technology described and claimed herein is not limited to or by the features or embodiments identified in this brief summary, which is included for purposes of illustration only and not restriction. Additional embodiments may be disclosed in the detailed description below.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIGS. 1A-1I. HSC-derived precursors are dispensable for osteoclasts and bone development. FIG. 1A: Top: Representative photographs of teeth of control littermates and Csf1Cre; Csf1rfl/fl 3-4 week old mice (right) and controls and Csf1rCre; Tnfrsf11afl/fl mice (left). Bottom left: Representative photographs of leg bones from controls and Csf1rCre; Tnfrsf11afl/fl mice, white arrowhead points to lack of blood cells. Right: bar graphs represent osteoclast numbers in E18.5, P7, and 4-week old Csf1rCre; Tnfrsf11afl/fl mice and littermates (n=3/3), and representative hematoxylin/TRAP staining of femur sections; arrow indicates an osteoclast. FIG. 1B: Representative photographs of teeth of 3-4 week old Flt3rCre; Tnfrsf11afl/fl Flt3Cre; Csf1rfl/fl; VavCre; Csf1rfl/fl and littermates. FIG. 1C: Bone marrow CD45+ cells numbers in Flt3rCre; Tnfrsf11afl/fl and littermates at 4 (n=4/3) and 22 week old (n=4/5) and VavCre; Csf1rfl/fl and littermates at 4 (n=8/8) and 62 week old (n=5/8) of the indicated age. FIG. 1D: Osteoclast counts in femurs from Flt3rCre; Tnfrsf11afl/fl; Flt3rCre; Csf1rfl/fl, VavCre; Csf1rfl/fl mice and littermates of the indicated ages. FIG. 1E: Representative hematoxylin/TRAP staining of femur sections from 22 week old Flt3rCre; Tnfrsf11afl/fl, 62 week old VavDD/Cre; Csf1rfl/fl mice and littermates. White arrowhead points to trabecular bone. FIG. 1F: Quantitative analysis of bone volume/total volume ratio of humerus or femurs from Flt3rCre; Tnfrsf11afl/fl (n=4), Flt3Cre; Csf1rfl/fl (n=5) and controls littermates (n=8), and VavCre; Csf1rfl/fl (n=5) mice and controls littermates (n=5) as determined by MicroCT and Statistical significance determined by ANOVA. FIG. 1G: Expression and MFI of YFP in in TRAP+ multinucleated cells from Csf1rCre; Rosa26LSL-YFP mice at embryonic day E16.5, E18.5, P7, and 6 months, (n=3 per time point). For MFI and percentages, at least 100 osteoclasts were quantified per time point, genotype, displayed as a box plot, min, Q1, median, Q3 and max. FIG. 1H as in FIG. 1G for Flt3Cre; Rosa26LSL-YFP mice at E16.5, E18.5, P7, 4 weeks (4w), 3 months, and 6 months. Data are mean±SD, circles represent individual independent biological replicates, n indicates number of mice per group. Statistical significance was analysed with GraphPad Prism using unpaired two-tailed t-tests unless otherwise indicated. FIG. 1I. Representative confocal microscopy image of frozen sections from mice in (FIGS. 1G, 1H). Statistical significance was analysed with GraphPad Prism using unpaired t-tests and two-way ANOVA with Tukey's multiple comparisons test as indicated in the figure legends. Significance was considered at p value (p) *p<0.05; **p<0.01; ***p<0.001; ****p<0.0001.
  • FIGS. 2A-2J. EMP derived osteoclasts are required for bone development. FIG. 2A: MGC number in femur anlage's ossification centers from E15.5/E16.5 Myb−/− (n=6) and littermate controls (n=3). FIG. 2B: Representative confocal microscopy pictures of frozen sections from FIG. 1A stained with fluorescent TRAP and TO-PRO-3. FIG. 2C: Percent of TRAP+ cells expressing YFP in femur anlage from E15.5 Csf1rMerCreMer; Rosa26LSL-YFP mice (n=8) and Cre negative controls (n=5), pulsed at E8.5 with 4-OHT. FIG. 2D: Representative confocal microscopy picture from FIG. 2C. FIG. 2E: Representative photographs of teeth of control littermates (Cre), Tnfrsf11aKoba-Cre; Csf1rfl/fl and Tnfrsf11aWask-Cre; Csf1rfl/fl mice (Cre+). FIG. 2F: Leg bones from Tnfrsf11aCre; Csf1rfl/fl mice (Cre+, n=6) and control littermates (Cre, n=6) at P7 and 4 weeks. Arrowhead points to the color of bone. FIG. 2G: Representative MicroCT scans of long bones from mice in FIG. 2E (n=6 per genotype). FIG. 2H: Skull length from 3 weeks old Csf1r−/− (n=6) controls littermates (n=12) and Tnfrsf11aWask-Cre; Csf1rfl/fl mice (n=4) as determined by MicroCT. FIG. 2I: Osteoclasts counts in bone sections from E18.5, P7, and 3-4 week old Tnfrsf11aKoba-Cre; Csf1rfl/fl mice and littermate controls. FIG. 2J: Numbers of bone marrow CD45+ cells determined by flow cytometry in 4 weeks old littermates (Cre, n=13,27), Tnfrsf11aKoba-Cre; Csf1rfl/fl (n=9) and Tnfrsf11aWask-Cre; Csf1rfl/fl (n=23) mice (Cre+). Data are mean±SD, circles on graphs represent individual mice, n indicates the number of mice per group. Statistical significance was analysed with GraphPad Prism using unpaired two tailed t-tests. Significance was considered at p value (p) *p<0.05; **p<0.01; ***p<0.001; ****p<0.0001.
  • FIGS. 3A-3G. In vivo dynamics of osteoclasts. FIG. 3A: Parabiosis of Csf1rCre; Rosa26LSL-YFP mouse surgically paired with a Csf1rCre; Rosa26LSL-tdTomato partner for 4 to 8 weeks. Representative (n=3) confocal microscopy image of frozen sections from the femur of a Csf1rCre; Rosa26LSL-YFP partner stained with antibodies for tdTomato (red) and YFP (green), ELF97 (blue) and TOPRO-3 (grey). FIG. 3B: Pie graphs represent the percentage of tdTomato positive (red), YFP positive (green) and double tdTomato/YFP positive cells (yellow) among BM mononuclear cells (MNC), Megakaryocytes (Mega) and Multinuclear Giant Cells (MGC), from parabionts paired for 1, 2, 3, 4, and 8 weeks (n=8). FIG. 3C: As in FIG. 3B for parabionts separated after 4 weeks and analysed 14 weeks and 24 weeks after separation (n=3). FIG. 3D: Scatter plots represent the Mean Fluorescent Intensity (MFI) of individual TRAP+ MGC for YFP (Y axis) and tdTomato (X axis), and histograms represent the overlayed distribution of the MFI values for YFP and tdTomato in TRAP+ MGC at the indicated time-points. FIG. 3E: Bar graph display number of nuclei per TRAP+ MGC in femurs from wild type mice at 1, 3, and 6 month of age (n=3 mice per time point). FIG. 3F: Representative confocal microscopy image of an EdU-labeled nucleus in a TRAP+ osteoclast, and histogram displaying the percentages of TRAP+ osteoclast with EdU-labeled nuclei and the number of labeled nuclei per cell 72 hrs after intravenous pulse-labeling with EdU (n=5 mice). FIG. 3G: A model for development and maintenance of osteoclast syncytia. Data are mean±SD, circles on graphs represent individual mice, n indicates the number of mice per group. Statistics: two-way ANOVA with Tukey's multiple comparisons test *p≤0.05 **p≤0.005.
  • FIGS. 4A-G. Rescue of Osteopetrosis. FIG. 4A: Bone volume over total volume (BV/TV %) of femurs from 10 week-old Cathepsin K−/− mice (n=3) after 6 weeks parabiosis with Cathepsin K+/− heterozygote mice and from positive (n=4) and negative (n=4) controls parabionts, analysed by Von Kossa staining. FIG. 4B: Monocyte transfer: histograms represent percentages of tdTomato+ cells among bone TRAP+ MGC from Csf1rCre; Rosa26LSL-YFP recipients analysed by confocal microscopy (left) 11 days (n=2) and 60 days (n=5) after i.v. transfer at age 6 week-old of 3×106 bone marrow Ly6C+ cells from Csf1rCre; Rosa26LSL-tdTomato donors, and percentages of tdTomato+ cells among bone marrow precursors and blood leukocytes analyzed by flow cytometry after 60 days (n=5, right). FIG. 4C: Representative high power confocal microscopy image of the femur of a recipient mouse 60 days after i.v. transfer (from FIG. 4B), stained with antibodies for tdTomato and YFP, and ELF97 and TOPRO-3. FIG. 4D: Representative photographs of teeth, and CT scan of leg bones from Csf1Cre; Csf1rfl/fl mice (n=3) transferred with monocytic cells from Csf1rCre; Rosa26LSL-YFP donors at post-natal day 5, 8, and 11, and from wt and non-transferred Csf1cre; Csf1rfl/fl controls. Arrows indicated the presences of teeth eruption (upper panel) and bone marrow cavity (lower panel). FIG. 4E: Representative confocal microscopy image of femur from mice #3 in FIG. 4D, stained with antibodies for YFP, ELF97 and TOPRO-3. FIG. 4F: Histograms represent (Left) the number of TRAP+ osteoclasts number in bone sections from mice in FIG. 4D and non-transferred controls and (Right) the percentage of TRAP+ cells that are YFP+ in transferred mice. Symbols represents individual mice, values are average for 3 sections per mice. At least n=100 osteoclasts were quantified per mice. FIG. 4G: Histograms represent percentages of YFP+ cells among bone marrow precursors and blood leukocytes in the recipient mice (in FIG. 4D) at the time of analysis. Data are mean±SD, circles on graphs represent individual mice, n indicates the number of mice per group. Statistical significance was analyzed with GraphPad Prism using ANOVA with Tukey's multiple comparisons test ***p<0.001; ****p<0.0001.
  • FIGS. 5A-G. Csf1rCre; Tnfrsf11afl/fl: Csf1rfl/fl; Csf1rCre, and Tnfrsf11aCre; Csf1rfl/fl mice are osteopetrotic. FIGS. 5A-B: Representative Computed Tomography scans (nanospect CT) of 4 week old Csf1rCre; Tnfrsf11afl/fl mice arrows point skull deformation, and absence of bone marrow cavity in mutant mice. FIG. 5C: H&TRAP staining of bone sections of 4 week old Csf1rCre; Tnfrsf11afl/fl mice showing closure of the bone marrow. Histology of paraffin sections (5 μm thickness) corroborates the phenotype from mice in FIG. 5B. FIG. 5D: Representative photograph of inguinal lymph nodes in Csf1rCre; Tnfrsf11afl/fl mice. FIG. 5E: Representative photograph of Csf1rCre; Tnfrsf11afl/fl and littermates controls. FIG. 5F: Representative computed tomography scan reconstructions (nanospect CT) of 4 weeks-old Tnfrsf11a(Koba)Cre; Csf1rfl/fl mice. Arrows point to skull deformation, but the presence of bone marrow cavity in mutant mice, as in FIGS. 5A, B. FIG. 5G: Representative Computed Tomography scans of Csf1Cre; Csf1rfl/fl mice. Arrows shows skull deformation, and absence of bone marrow cavity in mutant mice as in A,B. CT scans and photographs are representative of >10 litters.
  • FIGS. 6A-N. Bone histology and Flow cytometry analysis of bone marrow phenotypic KSL, LT-HSCs, ST-HSCs, and MPPs in mice of indicated genotypes. FIG. 6A: Young Flt3Cre; Csf1rfl/fl and Flt3Cre; Tnfrsf11afl/fl mice have normal long bone: H&TRAP staining of bone sections from 4 week old Flt3rCre; Tnfrsf11afl/fl mice showing normal bone structure and bone marrow cavity. FIG. 6B: LSK cell numbers in bone marrow Flt3rCre; Tnfrsf11afl/fl (n=3), Flt3rCre; Tnfrsf11afl/+ mice (n=2), and littermate controls (n=4) in 3-4 week old mice and Flt3rCre; Tnfrsf11afl/fl (n=5), Flt3rCre; Tnfrsf11afl/+ mice (n=4), and littermate controls (n=4) in 22 week old mice. FIG. 6C: H&TRAP staining of bone sections from 4 week old Flt3Cre; Csf1rfl/fl mice showing normal bone structure and bone marrow cavity. FIGS. 6D, E: Phenotypic long-term hematopoietic stem cells (LT-HSCs) are reduced in aged but not old VavCre; Csf1rfl/− mice, (FIGS. 6F, G) LT-HSC are reduced in in young Csf1r−/− and to a lesser extent in young Tnfrsf11a(Wask)Cre; Csf1rfl/− Numbers per 2 femur are shown. FIG. 6H: Flow cytometry analysis of F4/80+ cells in brain (microglia) and epidermis (Langerhans cells) in E18.5 Tnfrsf11aCre; Csf1rfl/fl embryos and littermate controls (n=3 per group). FIG. 6I: H&TRAP staining of bone sections from P7 Tnfrsf11aCre; Csf1rfl/fl and littermate control showing absence of the bone marrow cavity. FIG. 6J: Flow cytometry plot of Fetal liver at E15.5 (B/C representative results of 3 experiments). FIG. 6K: LSK numbers in bone marrow of 3-4 week old Tnfrsf11aCre(wask); Csf1rfl/fl (n=24) and littermate controls (n=20) and Tnfrsf11aCre(koba); Csf1rfl/fl (n=7), Tnfrsf11aCre(koba); Csf1rfl+l (n=6) and littermate controls (n=8). FIG. 6L: For comparison, LSK numbers in bone marrow of 3-4 week old Csf1r−/− (n=22) and littermate controls (n=21). FIG. 6M: Representative micrographs of femur sections from 4 week old Tnfrsf11aCre; Csf1rfl/fl and littermate control stained with hematoxylin/TRAP. FIG. 6N: Blood leukocytes numbers in 4 week old Tnfrsf11aCre; Csf1rfl/fl mice (n=5), Tnfrsf11aCre; Csf1rfl/+ mice (n=6) and littermate controls (n=12). Circles represent individual mice, results from 3 independent experiments. Data are mean±SD, circles on graphs represent individual mice, n indicates the number of mice per group. Statistical significance was analysed with GraphPad Prism using unpaired two tailed t-tests. Significance was considered at p value (p) *p<0.05; **p<0.005; ***p<0.0005; ****p<0.0001. LT-HSC: Lin− Kit+ Sca-1+; Kit+ Sca1+CD34− F1t3−. Phenotypic short-term hematopoietic stem cells (ST-HSC): Lin− Kit+ Sca-1+CD34+F1t3−. Multipotent progenitors (MPP): Lin− Kit+ Sca-1+CD34+F1t3+. Lin: CD3 CD19 NK1.1 Ter119 CD11b Gr1 B220.
  • FIGS. 7A-E. Bone histomorphometry in old Flt3rCre; Tnfrsf11afl/fl, Flt3Cre; Csf1rfl/fl and VavCre; Csf1rfl/fl mice and control littermates. FIG. 7A: Representative Micro CT pictures, genotype is indicated. FIG. 7B: Bone length, connectivity density (Conn density), Trabecular Number (Tb.N.), and Trabecular Spacing (Tb.Sp.) analyzed by microCT in aged Flt3rCre; Tnfrsf11afl/fl (n=4), Flt3Cre; Csf1rfl/fl (n=4) and control littermates (n=7). FIG. 7C: Bone histomorphometry as in FIG. 7B, in VavCre; Csf1rfl/fl mice and control littermates (n=5). FIGS. 7D, E: Dynamic bone histomorphometry in aged Flt3Cre; Tnfrsf11afl/fl and Flt3Cre; Csf1rfl/fl mice using in vivo calcein labeling. Representative micrographs of calcein labeling (green) of femura of mice from the indicated genotypes and ages (FIG. 7D). Scale bars represent 200 μm (top) and 50 μm (bottom). Quantification of calcein labeling by fluorescence microscopy of mineralized surface/bone surface (MS/B5), mineral apposition rate (MAR), and bone formation rate/bone surface (BFR/BS) in aged Flt3rCre; Tnfrsf11afl/fl (n=5), Flt3Cre; Csf1rfl/fl (n=3) and control littermates (n=10) Data are mean±SD, circles on graphs represent individual mice, n indicates the number of mice per group. Statistical significance was analysed with GraphPad Prism using unpaired two tailed t-tests. Significance was considered at p value (p) *p<0.05; **p<0.005; ***p<0.0005; ****p<0.0001.
  • FIGS. 8A-E. Colonization of the bone marrow by Csf1r+ and Flt3+ hematopoietic cells. FIG. 8A: Representative confocal microscopy images of frozen sections from Flt3Cre; Rosa26LSL-YFP and Csf1rCre; Rosa26LSL-YFP mice analyzed at E16.5 (n=3). FIG. 8B: YFP labeling efficiency in Flt3Cre; Rosa26LSL-YFP mice analyzed by flow cytometry in the indicated cell populations, and by confocal microscopy on frozen bone sections at the indicated time (Right). Insets show YFP expression on individual osteoclasts. YFP: anti-GFP antibody, TRAP: ELF97 fluorescent substrate, TO-PRO-3: nuclear stain. FIG. 8C: YFP labeling efficiency in Csf1rCre; Rosa26LSL-YFP mice analyzed as in FIG. 8A. Data from FIGS. 8A, B are representative of at least 3 experiments by time point and genotypes. Circles represent individual mice. Genetic lineage tracing of osteoclasts in ossification centers using Csf1rMeriCreMer; Rosa26LSL-YFP. Representative high power confocal microscopy images from embryonic femurs showing MGCs in primary ossification centers from Csf1rMeriCreMer; Rosa26LSL-YFP FIG. 8D: E18.5 embryos pulsed with 4-hydroxytamoxifen at E8.5 showing YFP expression in MGCs after Cre recombination and quantitated as MFI (right bar graph) from Cre+(n=8) and Cre−(n=4), and FIG. 8E unpulsed controls showing the lack of YFP in Cre+(n=4) and Cre−(n=4). Sections were labeled with antibodies against YFP, TRAP (ELF97 substrate) and TO-PRO-3 as a nuclear stain.
  • FIGS. 9A-D. Tnfrs11a(wask)Cre knock-in mice allow deletion of target genes in fetal macrophages, but not definitive HSC and their progeny in blood and tissues, while Vavcre mice allow deletion of target genes in definitive HSC, but not fetal macrophages. FIG. 9A: Bar graphs indicate percentages of eYFP expression by flow cytometry in cells from Tnfrsf11aCre; Rosa26LSL-YFP in the indicated cell types, organs, and time points. Data represent 3 independent experiments n numbers indicated on x axis. FIG. 9B: Lineage tracing in the fetal liver of Vavcre+; tdRFPwt/ki mice n numbers indicated on x axis. FIG. 9C: Representative molecular analysis of Csf1r deletion in purified bone marrow HSPC from 62 weeks old Vavcre; Csf1rfl/fl mice and controls (n=5). FIG. 9D: Representative photograph of teeth from 3 week old R.26-CreERT2+; Csf1rfl/− pulsed with Tamoxifen at E10.5 (n=3 mice from 3 independent litters). FL: Fetal Liver, HSPC: hematopoietic stem and progenitor cell, LT-HSC: Long-term Hematopoietic Stem Cells, MPP: Multipotent progenitors, PMN: polymorphonuclear cells, mono: monocytes, T: T cells, B: B cells, PEC: peritoneal exudate cells. Data are mean±SD, circles on graphs represent individual mice, n indicates the number of mice per group.
  • FIGS. 10A-F. Bone morphometric and dynamic histomorphometry effects of the deletion of Csf1r in P21 Tnfrsf11aCre; Csf1fl/fl mice. Bone volume/total volume (BV/TV, FIG. 10A), Bone length (FIG. 10B), connectivity density (Conn density, FIG. 10C), and Trabecular Number (Tb.N., FIG. 10D) was analyzed by microCT in 21 day/old mice. Csf1r−/−(n=4) and control littermates (n=7), Tnfrsf11a(wask)Cre+; Csf1rfll− (n=8) and Tnfrsf11a(wask)Cre+; Csf1rfll+(n=7), Tnfrsf11a(wask)Cre+; Csf1rfll−(n=3) and control littermates (n=5). FIG. 10E: Representative micrographs of calcein labeling (green) of femur of mice from the indicated genotypes and ages (n=4). Scale bars represent 50 μm. FIG. 10F: Quantification of calcein labeling by fluorescence microscopy: mineralized surface/bone surface (MS/BS), mineral apposition rate (MAR), and bone formation rate/bone surface (BFR/BS) in Tnfrsf11a(wask)Cre+; Csf1rfll− Csf1r−/− (n=4) and control littermates (n=15). Data are mean±SD, circles on graphs represent individual mice, n indicates the number of mice per group. For statistical analysis unpaired two-tailed t-test. *p≤0.05, **p≤0.005, ***p≤0.0005, ****p≤0.0001.
  • FIGS. 11A-C. EdU labeling of bone marrow myeloid cells. FIG. 11A: Short term kinetics: EdU (20 μg/g) is injected intraperitoneally to C57B16/N mice at t=0 min. Mice are sacrificed at the indicated time points and the % of EdU+ cells (blue) and the geometric MFI of EdU+ cells (red) and determined by flow cytometry, showing rapid EdU incorporation. % of EdU+ cells plateau at ˜30 min, and gMFI plateau at ˜75 minutes. Following a first round of cell division, ˜50% of monocytic cells are labeled after 8 to 12 hrs (n=3-8 mice see source data). FIG. 11B: Long term kinetics: (1-240 hrs) EdU is injected as in FIG. 11A and % of EdU+ monocytic cells in bone marrow (top) and blood (bottom) is determined by flow cytometry, showing that labeling of ˜50% of monocytic cells is observed for ˜2 days. Labeled cells are not detectable after 3 days. Circles represent determination from individual mice, data for each time point are pooled from 2-3 independent experiments see source data. FIG. 11C: Parabiosis between Csf1rCre; Rosa26LSL-YFP and Csf1rCre; Rosa26LSL-tdTomato pairs as described in (FIG. 3), paired for 1-8 weeks, and from Csf1rCre; Rosa26LSL-YFP partners separated after 4 weeks of parabiosis and analyzed 4 weeks, 14 weeks and 24 weeks after separation. Scatter plots represent the Mean Fluorescent Intensity (MFI) of individual TRAP+ MGC for YFP (Y axis) and tdTomato (X axis), and histograms represent the overlaid distribution of the MFI values for YFP and tdTomato in TRAP+ MGC at the indicated time-points. Data are mean±SD, circles on graphs represent individual mice, n indicates the number of mice per group.
  • FIGS. 12A-C. FACS analysis of monocyte purification, and blood/bone marrow from transferred Csf1rCre; Csf1rfl/fl mice. FIG. 12A: Representative flow cytometry plots of purified bone marrow monocytes from magnetic bead based enrichment (MACS), percentage of live YFP+ monocytes is indicated. Representative flow cytometry plots from blood (FIG. 12B) and bone marrow (FIG. 12C) of 14 day old mice transferred with 1E6 YFP+ monocytes on day 5,8,11, the percentage of YFP+ cells is indicated. Experiments A-C are representative of 3 independent experiments.
  • FIG. 13. Rescue of osteoclasts by monocyte transfer in Csf1Cre; Csf1rfl/fl mice. High power confocal microscopy images of frozen sections from Csf1Cre; Csf1rfl/fl mice transferred with monocytes from Csf1rCre Rosa26LSL-YFP and controls, stained with antibodies for YFP, TRAP substrate ELF97 and TOPRO-3 as a nuclear stain. Examples of multinucleated Trap+, YFP+ cells (osteoclasts) are indicated with dotted lines. n=3 mice from independent litters. Numbers (#1, 2, 3) correspond to the mice in FIG. 4D.
  • FIGS. 14A-F. Csf1r deletion in Tnfrsf11aCre Csf1rF/F mice results in lack of tissue macrophages and osteoclasts at birth, while definitive HSC and their progeny are present in the fetal liver and blood. FIG. 14A: Flow cytometry analysis of F4/80+ cells in brain (microglia) and epidermis (Langerhans cells) in E18.5 Tnfrsf11aCre+; Csf1rF/F embryos and littermate controls. FIG. 14B: H&TRAP staining of bone sections from P7 Tnfrsf11aCre+; Csf1rF/F and Tnfrsf11aCre−; Csf1rF/F littermate control showing absence of the bone marrow cavity. FIG. 14C: Flow cytometry plot of Fetal liver at E15.5. FIG. 14D LSK numbers in bone marrow of 3-4 week old Tnfrsf11aCre+; Csf1rF/F and littermate controls. FIG. 14E: For comparison, LSK numbers in bone marrow of 3-4 week old Csf1r−/− and littermate controls. FIG. 14F: Blood leukocytes numbers in 4 week old Tnfrsf11aCre+; Csf1rF/F mice and littermate controls. Circles represent individual mice, results from 3 independent experiments.
  • FIGS. 15A-D. Bone morphometric effects of the deletion of Csf1r in P21 Tnfrsf11aCre+; Csf1rF/− or F/F mice. Bone volume/total volume (BV/TV, FIG. 15A), Bone length FIG. 15B, connectivity density (Conn density, FIG. 15C), and Trabecular Number (Tb.N., FIG. 15D) was analyzed by microCT in 21 day/old mice of indicated genotype. Controls (ctrls) included Tnfrsf11a(wask)Cre+; Csf1rF/+ or Tnfrsf11a(koba)Cre−; Csf1rF/F and Tnfrsf11a(koba)Cre+; Csf1rF/+ types. Circles represent 1 mouse with 3-7 mice per group. For statistical analysis unpaired two-tailed t-test. *p≤0.05, **p≤0.005, ***p≤0.0005, ****p≤0.0001.
  • FIGS. 16A-B. EdU labeling of bone marrow myeloid cells. FIG. 16A: Short term kinetics: EdU (20 μg/g) is injected intraperitoneally to C57B16/N mice at t=0 min. Mice are sacrificed at the indicated time points and the % of EdU+ cells (blue) and the geometric MFI of EdU+ cells (red) and determined by flow cytometry, showing rapid EdU incorporation. % of EdU+ cells plateau at ˜30 min, and gMFI plateau at ˜75 minutes. Following a first round of cell division, ˜50% of monocytic cells are labeled after 8 to 12 hrs. FIG. 16B: Long term kinetics: (1-240 hrs) EdU is injected as in (A) and % of EdU+ monocytic cells in bone marrow (top) and blood (bottom) is determined by flow cytometry, showing that labeling of ˜50% of monocytic cells is observed for ˜2 days. Labeled cells are not detectable after 3 days. Circles represent determination from individual mice, data for each time point are pooled from 2-3 independent experiments.
  • DETAILED DESCRIPTION
  • It is to be appreciated that certain aspects, modes, embodiments, variations and features of the present technology are described below in various levels of detail in order to provide a substantial understanding of the present technology. The definitions of certain terms as used in this specification are provided below. Unless defined otherwise, all technical and scientific terms used herein generally have the same meaning as commonly understood by one of ordinary skill in the art to which this present technology belongs.
  • I. Definitions
  • The following terms are used herein, the definitions of which are provided for guidance.
  • As used herein, the singular forms “a”, “an” and “the” include plural referents unless the content clearly dictates otherwise. For example, reference to “a cell” includes a combination of two or more cells, and the like.
  • As used herein, the term “about” and the use of ranges in general, whether or not qualified by the term about, means that the number comprehended is not limited to the exact number set forth herein, and is intended to refer to ranges substantially within the quoted range while not departing from the scope of the invention. As used herein, “about” will be understood by persons of ordinary skill in the art and will vary to some extent on the context in which it is used. If there are uses of the term which are not clear to persons of ordinary skill in the art given the context in which it is used, “about” will mean up to plus or minus 10% of the particular term.
  • A “chimeric nucleic acid” comprises a coding sequence or fragment thereof linked to a nucleotide sequence that is different from the nucleotide sequence with which it is associated in cells in which the coding sequence occurs naturally.
  • As used herein, the terms “effective amount,” or “therapeutically effective amount,” and “pharmaceutically effective amount” refer to a quantity sufficient to achieve a desired therapeutic and/or prophylactic effect, e.g., an amount which results in the prevention of a disease, condition, and/or symptom(s) thereof. In the context of therapeutic or prophylactic applications, the amount of a composition administered to the subject will depend on the type and severity of the disease and on the characteristics of the individual, such as general health, age, sex, body weight, and tolerance to the composition drugs. It will also depend on the degree, severity, and type of disease or condition. The skilled artisan will be able to determine appropriate dosages depending on these and other factors. In some embodiments, multiple doses are administered. Additionally or alternatively, in some embodiments, multiple therapeutic compositions or compounds are administered. In the methods described herein, compositions comprising the monocytic cells of the present technology, may be administered to a subject having one or more signs, symptoms, or risk factors of osteopetrosis, including, but not limited to stunted growth, skeletal deformity, increased likelihood of bone fracture, anemia, recurrent infections, hepatosplenomegaly, facial paralysis, abnormal cortical bone morphology, abnormal form of vertebral bodies, abnormal temperature regulation, abnormality of the ribs, abnormality of vertebral epiphysis morphology, bone pain, cranial nerve paralysis, craniosynostosis, hearing impairment, and hypocalcemia. For example, a “therapeutically effective amount” of the compositions of the present technology, includes levels at which the presence, frequency, or severity of one or more signs, symptoms, or risk factors of osteopetrosis are, at a minimum, ameliorated. In some embodiments, a therapeutically effective amount reduces or ameliorates the physiological effects of osteopetrosis, and/or the risk factors of osteopetrosis, and/or the likelihood of developing osteotpetrosis. In some embodiments, a therapeutically effective amount is achieved by multiple administrations. In some embodiments, a therapeutically effective amount is achieved with a single administration.
  • The term “engineered” is used herein to refer to a cell or organism that has been manipulated to be genetically altered, modified, or changed, e.g., by disruption of the genome. For example, an “engineered monocytic cell” refers to a monocytic cell that has been manipulated to be genetically altered, modified, or changed. For example, in some embodiments, an engineered monocytic cell refers to a monocytic cell that has been transduced with a lentivirus designed to express a nucleotide sequence of interest, e.g., a cDNA coding for the wild type allele of any one or more of CA2, CLCN7, CTSK, CSF1R, IKBKG, ITGB3, OSTM1, PLEKHM1, TCIRG1, TNFRSF11A, or TNFSF11 under a strong promoter.
  • “Heterologous nucleic acid” refers to a nucleic acid, DNA, or RNA, which has been introduced into a cell (or the cell's ancestor), and which is not a copy of a sequence naturally found in the cell into which it is introduced. Such heterologous nucleic acid may comprise segments that are a copy of a sequence that is naturally found in the cell into which it has been introduced, or fragments thereof.
  • As used herein, “prevention,” “prevent,” or “preventing” of a disorder or condition refers to, in a statistical sample, reduction in the occurrence or recurrence of the disorder or condition in treated subjects/samples relative to an untreated controls, or refers delays the onset of one or more symptoms of the disorder or condition relative to the untreated controls.
  • As used herein “subject” and “patient” are used interchangeably and refer to a mammalian subject. In some embodiments, “subject” means any animal (mammalian, human, or other) patient that can be afflicted with osteopetrosis and when thus afflicted is in need of treatment. In some embodiments, the subject is a human.
  • “Treating,” “treat,” “treated,” or “treatment” of a disease or disorder includes: (i) inhibiting the disease or disorder, i.e., arresting its development; (ii) relieving the disease or disorder, i.e., causing its regression; (iii) slowing progression of the disorder; and/or (iv) inhibiting, relieving, or slowing progression of one or more symptoms of the disease or disorder.
  • It is to be appreciated that the various modes of treatment or prevention of medical diseases and conditions as described are intended to mean “substantial,” which includes total but also less than total treatment or prevention, and wherein some biologically or medically relevant result is achieved.
  • II. General
  • Described herein are compositions and methods for the treatment of osteopetrosis. Taken together, the data described herein identify: (i) the developmental origin of osteoclasts, the cells that shape bone architecture; (ii) a mechanism that controls the maintenance of osteoclast function after birth; and (iii) a novel strategy to treat osteopetrosis and to modulate osteoclast activity in vivo. In particular, the data described herein demonstrate that parabiosis or transfusion of monocytic cells results in long-term gene transfer in osteoclasts in the absence of HSC chimerism and can rescue an adult-onset osteopetrotic phenotype caused by cathepsin-K deficiency. Transfusion of monocytic cells is also sufficient to rescue bone development in early-onset autosomal recessive osteopetrosis in newborn mice.
  • III. Osteopetrosis
  • Osteopetroses are a heterogeneous group of genetic disorders characterized by increased bone density due to impaired bone resorption by osteoclasts. The increased bone density places the affected individual at an increased risk for bone fracture. Normally, bone growth is a balance between osteoblasts (cells that create bone tissue) and osteoclasts (cells that destroy bone tissue). Individuals with osteopetrosis have a deficiency of osteoclasts, resulting in too little bone resorption and too much bone creation. The types of osteopetrosis are distinguished based on their pattern of inheritance: autosomal dominant, autosomal recessive, or X-linked.
  • The signs and symptoms of osteopetrosis may vary depending on the type of the disease, and mild forms of osteopetrosis may be asymptomatic. However, the typical signs and symptoms of osteopetrosis include: stunted growth, skeletal deformity, increased likelihood of bone fracture, anemia, recurrent infections, hepatosplenomegaly, facial paralysis, abnormal cortical bone morphology, abnormal form of vertebral bodies, abnormal temperature regulation, abnormality of the ribs, abnormality of vertebral epiphysis morphology, bone pain, cranial nerve paralysis, craniosynostosis, hearing impairment, and hypocalcemia.
  • The signs and symptoms of autosomal dominant osteopetrosis (ADO; also known as Albers-Schonberg disease), include multiple bone fractures, scoliosis, arthritis in the hips, and/or osteomyelitis. Autsomal recessive osteopetrosis (ARO) is often characterized by one or more of a high risk of bone fracture resulting from minor bumps or falls, pinched nerves in the head and face, impaired bone marrow function, slow growth, short stature, dental abnormalities, hepatosplenomegaly, intellectual disability, and epilepsy. Individuals diagnosed with intermediate autosomal osteopetrosis (IAO), which is a form of osteopetrosis that can have either an autosomal dominant or recessive pattern or inheritance, may be characterized by one or more of a high risk of bone fracture and anemia, calcifications in the brain, intellectual disability, and renal tubular acidosis. Individuals with the X-linked pattern of inheritance may be characterized by one or more of lymphedema, anhidrotic ectodermal dysplasia, and immunodeficiency.
  • Mutations in several genes have been linked to the various forms of osteopetrosis or may underlie the development of osteopetrosis. These include mutations in CA2, CLCN7, CTSK, CSF1R, IKBKG, ITGB3, OSTM1, PLEKHM1, TCIRG1, TNFRSF11A (which encodes for receptor activator of NF-κB (RANK)), and TNFSF11 (which encodes for receptor activator of NF-κB ligand (RANKL)). Many of the genes associated with osteopetrosis are involved in the formation, development, and function of osteoclasts.
  • IV. Therapeutic Applications
  • In some embodiments, a method for treating or preventing osteopetrosis in a subject in need thereof, comprising administering a composition comprising a therapeutically effective amount of monocytic cells from a healthy donor to the subject is provided. In some embodiments, the subject is characterized by decreased expression of one or more genes implicated in or potentially underlying the development of osteopetrosis, such as CA2, CLCN7, CTSK, CSF1R, IKBKG, ITGB3, OSTM1, PLEKHM1, TCIRG1, TNFRSF11A, and TNFSF11. In some embodiments, the subject is characterized by a cathepsin K deficiency.
  • In some embodiments, a method for treating or preventing osteopetrosis in a subject in need thereof, comprising administering to the subject a composition comprising a therapeutically effective amount of monocytic cells engineered to express one or more genes implicated in or potentially underlying the development of osteopetrosis, such as CA2, CLCN7, CTSK, CSF1R, IKBKG, ITGB3, OSTM1, PLEKHM1, TCIRG1, TNFRSF11A, and TNFSF11 is provided. In some embodiments monocytic cells used for treatment are genetically modified to correct a genetic abnormality or to improve or changed cellular functioning according to known genetic engineering protocols. In some embodiments, a method of treating or preventing osteopetrosis in a subject comprises: (a) obtaining a sample of monocytic cells from the subject; (b) genetically correcting one or more mutations in the monocytic cells, (c) culturing the monocytic cells; and (d) providing the corrected monocytic cells to the subject. In some embodiments, a method for treating or preventing osteopetrosis comprises: (a) obtaining a sample of monocytic cells from the subject; (b) genetically engineering the cells to express a nucleotide sequence coding for the wild type allele of any one or more of CA2, CLCN7, CTSK, CSF1R, IKBKG, ITGB3, OSTM1, PLEKHM1, TCIRG1, TNFRSF11A, and TNFSF11 with a suitable transducing vector, such as a lentiviral vector; (c) culturing the engineered monocytic cells under conditions sufficient to express the nucleotide sequence; (d) removing the viral particles from the engineered monocytic cells; and (e) providing the engineered monocytic cells to the subject. In some embodiments, the transducing vector encoding the one or more of CA2, CLCN7, CTSK, CSF1R, IKBKG, ITGB3, OSTM1, PLEKHM1, TCIRG1, TNFRSF11A, and TNFSF11 is a mammalian expression vector. In some embodiments, the mammalian expression vector is a lentiviral vector or transposon vector.
  • Subjects suffering from osteopetrosis can be identified by any or a combination of diagnostic or prognostic assays known in the art. For example, typical symptoms of osteopetrosis include, but are not limited to, stunted growth, skeletal deformity, increased likelihood of bone fracture, anemia, recurrent infections, hepatosplenomegaly, facial paralysis, abnormal cortical bone morphology, abnormal form of vertebral bodies, abnormal temperature regulation, abnormality of the ribs, abnormality of vertebral epiphysis morphology, bone pain, cranial nerve paralysis, craniosynostosis, hearing impairment, and hypocalcemia.
  • For therapeutic applications, a composition comprising a therapeutically effective amount of monocytic cells from a health donor and/or monocytic cells engineered to express one or more genes selected from CA2, CLCN7, CTSK, CSF1R, IKBKG, ITGB3, OSTM1, PLEKHM1, TCIRG1, TNFRSF11A, and TNFSF11 is administered to the subject. In some embodiments, the composition is administered according to any acceptable transfusion regimen. In some embodiments, the composition is administered one, two, three, four, or five times per day. In some embodiments, the composition is administered more than five times per day. Additionally or alternatively, in some embodiments, the composition is administered every day, every other day, every third day, every fourth day, every fifth day, or every sixth day. In some embodiments, the composition is administered weekly, bi-weekly, tri-weekly, or monthly. In some embodiments, the composition is administered for a period of one, two, three, four, or five weeks. In some embodiments, the composition is administered for six weeks or more. In some embodiments, the composition is administered for twelve weeks or more. In some embodiments, the composition is administered for a period of less than one year. In some embodiments, the composition is administered for a period of more than one year or until a desired therapeutic outcome is observed in the subject.
  • In some embodiments, treatment of subjects diagnosed with or suspected of having osteopetrosis with one or more compositions of the present technology ameliorates or eliminates one or more of the following symptoms of osteopetrosis: stunted growth, skeletal deformity, increased likelihood of bone fracture, anemia, recurrent infections, hepatosplenomegaly, facial paralysis, abnormal cortical bone morphology, abnormal form of vertebral bodies, abnormal temperature regulation, abnormality of the ribs, abnormality of vertebral epiphysis morphology, bone pain, cranial nerve paralysis, craniosynostosis, hearing impairment, and hypocalcemia.
  • Prophylactic Methods
  • In one aspect, the present technology provides a method for preventing or delaying the onset of osteopetrosis or one or more symptoms of osteopetrosis in a subject at risk of having or developing osteopetrosis. In prophylactic applications, compositions of the present technology are administered to a subject susceptible to, or otherwise at risk of for osteopetrosis in an amount sufficient to eliminate or reduce the risk, or delay the onset of the disease, including biochemical, histologic and/or behavioral symptoms of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease.
  • Administration of a prophylactic compositions can occur prior to the manifestation of symptoms characteristic of the disease or disorder, such that the disease or disorder is prevented or, alternatively, delayed in its progression.
  • Examples
  • The following examples are provided by way of illustration only and not by way of limitation. Those of skill in the art will readily recognize a variety of non-critical parameters that could be changed or modified to yield essentially the same or similar results. The examples should in no way be construed as limiting the scope of the present technology, as defined by the appended claims.
  • Materials and Methods
  • Csf1riCre, Csf1rMeriCreMer, Csf1rflox were kindly provided by Dr Jeffrey Pollard, Csf1r−/− from Richard Stanley, Flt3Cre were kindly provided by Dr Thomas Boehm, Myb+/− mice were kindly provided by Dr John Frampton, and VavCre were kindly provided from Dr Thomas Graf. Tnfrsf11aflox mice were kindly provided by J M Penninger, Tnfrsf11akoba/Cre mice were kindly provided by Dr Yasuhiro Kobayashi, Ctsktm1(cre)Ska mice were kindly provided by Dr. Ostrowsky (MSKCC) and Tnfrsf11aWask/Cre mice were genereated in the Waskow Lab. Rosa.26-CreERT2 (R26-CreERT2) were kindly provided from Drs. Pierre Chambon and Anton Berns. Rosa26LSL-YEP (stock number: 006148) and Rosa26LSL-tdTomato (stock number: 007908) reporter mice were purchased from The Jackson Laboratory.
  • Animal procedures. Mice were bred and kept under specific pathogen conditions in separated ventilated cages in the animal facility of MSKCC and the Medical Theoretical Center of the TU Dresden. All experiments with osteopetrotic mice that lack teeth were performed with mice maximal 4 weeks of age that were kept with the lactating mother or provided with DietGel 76A (Clear H2O, 72-07-5022) to avoid secondary effects from malnutrition. Experiments were performed in adherence to the Institutional Review Board (IACUC 15-04-006) from MSKCC and Landesdirektion Dresden and were in compliance with relevant ethical regulations. Mice greater than 7 days old were sacrificed by cervical dislocation (TU Dresden), CO2 asphyxiation or anesthesia (MSKCC). To harvest embryos, pregnant females were sacrificed and embryos were collected by postmortem cesarean from the uterus and exsanguinated through decapitation in cold PBS (Fisher, 14190).
  • Genotyping: PCR genotyping was performed according to protocols described previously and indicated in Table 1. The investigators were not blinded to allocation during experiments and outcome assessment.
  • Primer 1 Primer 2 Final
    Mouse sequence sequence De- Anneal- Elonga- Elonga- Expected
    line 5′ -> 3′ 5′ -> 3′ naturation ing tion Cycles tion bunds
    CsflriCre Mutant TCTCT CTCTG 94° C.- 60° C.- 72° C.- 30 72° C.- Mutant:
    allele GCCCA ACAGA 45 sec 45 sec 45 sec 5 min 400 bp
    GAGTC TGCCA
    ATCCT GGACA
    CsflrMeriCreMer WT TCGAA TCTCT 94° C.- 60° C.- 72° C.- 30 72° C.- WT: 
    allele GCTTG GCCCA 45 sec 90 sec 90 sec 5 min 1 Kb
    CATGC GAGTC
    CTGCA ATCCT
    Mutant TCATT GATCG Mutant:
    allele CCAGA TGTTG 2 Kb
    ACCAG GGGAA
    AGO GCC
    Csflrflox Mutant GCCAC ACCCA 94°C- 60° C.- 72° C.- 35 72° C.- WT:
    allele CATGT GAGCC 30 sec 60 sec 60 sec 2 min 450 bp
    GTCCG CCCAC Floxed:
    TGCTT AGATA 500 bp
    Csflr−/− Mutant GGTGG CGTTT 95°C - 62° C.- 72° C.- 35 72° C.- Mutant:
    allele ATGTG CTTGT 20 sec 20 sec 40 sec 5 min 314
    GAATG GGTCA
    TGTGC GGGTG
    G C
    Flt3Cre Mutant ACGGA GAAGC 94° C.- 58° C.- 68° C.- 44 72° C.- Mutant:
    allele GTCCA ATGTT 30 sec 35 sec 60 sec 2 min 400 bp
    GGCAA TAGCT
    CTTCC GGCCC
    Myb+/− WT CCATG GTGCT 94° C.- 60° C.- 72° C.- 35 72° C.- WT:
    allele CGTCG TCGGC 30 sec 30 sec 60 sec 7 min 200 bp
    CAAGG GATGT
    TGGAA GGTAA
    C
    Mutant CCATG TGGCC Mutant:
    allele CGTCG GCTTTT 300 bp
    CAAGG CTGGA
    TGGAA TTCAT
    C C
    Rosa.26CreERT2 Mutant GCCTG AATGG 95°C - 67° C.- 72° C.- 40 72° C.- Mutant:
    allele CATTA TGTTG 20 sec 20 sec 50 sec 2 min 700 bp
    CCGGT CCGCG
    CGATG CCATC
    CAACG TGCCA
    A C
    Rosa26LSL-YFP WT AAGTC GGAGC 94° C.- 60° C.- 72° C.- 34 72° C.- WT:
    allele GCTCT GGGAG 40 sec 60 sec 60 sec 5 min 525 bp
    GAGTT AAATG
    GTTAT GATAT
    G
    Mutant AAGTC GCGAA Mutant:
    allele GCTCT GAGTT 300 bp
    GAGTT TGTCC
    GTTAT TCAAC
    C
    Rosa26LSL-tdfTomato WT AAGGG CCGAA Step 1: 65° C.- 68° C.- 10 72° C.- WT: 
    alelle AGCTG AATCT 94° C.- 15 sec 10 sec 2 min 297 bp
    CAGTG GTGGG 20 sec (0.5 C Mutant:
    GAGTA AAGTC decrease 196 bp
    per
    cycle)
    Mutant GGCAT CTGTT Step 2: 60° C.- 72° C.- 28
    allele TAAAG CCTGT 94° C.- 15 sec 10 sec
    CAGCG ACGGC 15 sec
    TATCC ATGG
    Tnfrsfllaflox WT AGTGT CTGGT 94° C.- 60° C.- 72° C.- 35 72° C.- WT:
    alelle GCCTG GGTTG 30 sec 30 sec 40 sec 5 min 256 bp
    GCATG TTCTCC Floxed:
    TGCAG TGGTG 390 bp
    ACCTT TCAT Delta:
    Mutant AGTGT GGCAG 566 bp
    allele GCCTG AACTC
    GCATG GGATG
    TGCAG CACAG
    ACCTT ATTGG
    TnfrsfllaKoba/Cre WT TGAAG AATAG 94° C.- 60° C. - 72° C.- 35 72° C.- WT:
    allele GGTGA GGGTG 30 sec 30 sec 30 sec 5 min 530 bp
    CATCA GGGTG Mutant:
    TCGTG ATA 274 bp
    GT
    Mutant ACTTC AATAG
    allele TCCAT GGGTG
    GGTAG GGGTG
    CCTCC ATA
    TnfrsfllaWask/Cre Mutant AACCT GTCAA 95° C.- 58° C.- 68° C.- 35 72° C.- Mutant:
    allele GAGGA AGTCA 30 sec 30 sec 60 sec 2 min 210 bp
    TGTGA GTGCG
    GGGAC TTCAA
    TA AG
    VavDD/Cre Mutant GCCTG AATGG 95° C.- 67° C. - 72° C.- 40 72° C.- Mutant:
    allele CATTA TGTTG 20 sec 20 sec 50 sec 2 min 700 bp
    CCGGT CCGCG
    CGATG CCATC
    CAACG TGCCA
    A C
  • Fate mapping with tamoxifen inducible Cre models. For timing of embryonic development, mice were crossed at night, the following day a positive vaginal plug was considered as 0.5 days post-coitum (dpc), as previously described. Embryos were harvested as indicated below. For postnatal time point, pregnant females were monitored for the date of delivery, caesarean sections were carried out at term and neonates were fostered using lactating CD-1 females.
  • Csf1rMeriCreMer female mice were crossed with male Rosa26LSL-YFP/LSL-YFP mice. Cre recombination in Csf1rMeriCreMer; Rosa26LSL-YFP embryos at embryonic day E8.5 was induced with a single dose of 4-hydroxytamoxifen (4-OHT) injected intraperitoneally in pregnant mothers at a dose of 75 μg/g of body weight supplemented with 37.5 μg/g of progesterone as previously described. Cre-mediated recombination in Rosa.26-CreERT2 tamoxifen (TAM) was introduced to pregnant mother by a single TAM gavage (5 mg) at E10.5, supplemented with progesterone (37.5 μg/g body weight resolved in Sunflower seed oil, Sigma-Aldrich) was injected i.p. directly after gavage. To analyze newborn or 3 weeks old mice, caesarean sections were carried out at term and neonates were fostered using lactating CD-1 females.
  • Parabiosis. For cellular complementation female Csf1iCre mice were crossed to male Rosa26LSL-YFP/YFP or Rosa26LSL-tdTomato/tdTomato mice. Csf1riCre; Rosa26LSL-YFP and Csf1riCre; Rosa26LSL-tdTomato females were used for parabiosis. For rescue of CathepsinK activity 4 week old female CtskCre/Cre and control littermate mice were used for parabiosis. Parabionts were kept on Sulfamethoxazole/Trimethoprim (Sulfatrim) diet for up to 8 weeks. Ex-parabionts were separated after 4 weeks for cellular complementation and 6 weeks for rescue of CathepsinK activity.
  • Surgical procedure, pre-operative procedure: weight-matched female partner mice for parabiosis were caged together few days before surgery. One day before the surgery the fur from lateral sides of mice was carefully removed with a trimmer followed by depilatory cream (for 3 minutes) at the site of surgery, excess of fur was removed with a moist gauze pad. Left side partner is shaved on the right side and vice versa. This procedure was performed under Isofluorane inhalation anesthesia. Mice were fed with food supplemented with Sulfatrim ad libitum one day prior to surgery.
  • Surgery: Mice were anesthetized intraperitoneally with 150 mg/kg of ketamine and 15 mg/kg of xylazine. Sterile eye lubricant (Paralube Vet Ointment, 17033-211-38) was applied to both eyes to prevent corneal drying during surgery. Following confirmation that a suitable anesthetic plane (no response to stimulation) has been attained, mice were placed in a supine position on a surgical tray with heat support provided by a heating pad. The surgical site was cleaned 3 times with cotton swabs soaked in povidone-iodine (Betadine) then with 70% ethanol. Before surgery a volume of 0.2 ml of anesthetic agent bupivacaine (Marcaine 0.25-0.5% solution) was applied locally. Surgery was performed by a longitudinal skin incision on the lateral side of mice, approximately 0.5 cm above the elbow to 0.5 cm below the knee joint. Mice were laid side-by-side in close contact and the ligaments of the two knees and elbows were sutured together using monofilament non-absorbable suture. Then, the skin incisions was closed by apposing and clipping skin to skin of the pair with 9 mm wound clips.
  • Post-operative procedure: immediately after surgery mice were injected subcutaneously with 2 mg/kg of meloxicam and 0.5 mg/kg of buprenorphine and for a maximum of 48 hours postoperatively. Mice are provided with Sulfatrim and Hydrogel (Clear H2O, 70-01-5022) in medicups. Wound clips were removed 14 days after surgery under Isofluorane anesthesia.
  • Parabiosis separation procedure: Mice were anesthetized and prepared for surgery as indicated above. Mice were separated at the site of parabiosis junction. Using scissors, the skin joining both mice is cut longitudinally. The sutures around the elbows and knees are cut and removed. The resulting wound is closed with 9 mm wound clips. Mice were injected subcutaneously with 2 mg/kg of meloxicam and 0.5 mg/kg of buprenorphine and for a maximum of 48 hours postoperatively. Wound clips were removed 14 days after surgery.
  • Analysis: Bones were prepared for histology on frozen sections as detailed below, and stained with antibodies to detect fluorescent proteins: anti-GFP biotin, anti-RFP (Abcam), fluorescent TRAP staining and TO-PRO-3 as a nuclear stain. Histological sections of 15 μm thickness were scanned by confocal microscopy at 1.5 μm Z stacks. Sections were quantified for the number of TRAP+ multinuclear cells (more than 3 nuclei per cell) and their YFP and Tomato expression using Imaris in 3D view and individual Z stacks. Pictures of the region of interest (area=2 mm2) were then generated in Tiff format and analyzed by ImageJ software using ROI manager to calculate the mean fluorescence intensity (MFI) of YFP and Tomato for individual osteoclasts. For rescue of CathepsinK activity, dissected femurs were fixed in 10% neutral buffered formalin for 24 hours. Undecalcified bones were embedded in methyl methacrylate resin, and 7-μm sections were prepared on a rotation microtome. For mineralized bone volume over the total volume % (BV/TV %), sections were de-plastified and stained with von Kossa reagent (1% Silver nitrate/Sodium formamide/5% Sodium thiosulfate) counterstained with Van Gieson solution.
  • EdU pulse labeling. 12 week old C57BL/6N mice (Charles River) were injected intraperitoneally with 25 μg/g of a 2.5 mg/mL solution of EdU prepared extemporaneously (Fisher C10420, Click-iT EdU Alexa Fluor 488 Flow Cytometry Assay Kit). Blood, bone marrow, and bone samples were collected at 60 hrs and 72 hrs post injection and bone were prepared for histology of frozen sections as indicated above for adult mice.
  • Histology of frozen sections. Frozen sections were cut at 15 μm thickness using cryofilm and stained overnight with rat anti-Tubulin (Abcam, ab6160, 1:200) and with secondary goat anti-rat Alexa Fluor 555 (ThermoFisher Scientific A21430), as described below. Sections were washed 3 times with PBS and stained for EdU using Click-iT EdU Alexa Fluor 488 Imaging Assay Kit (Thermo Fisher Scientific, C10337). Click-it reaction: 357.5 μL of 1× Click-it reaction buffer, 40 μL of CuSO4 solution (100 mM), 2.5 μL of Alexa Fluor 488 azide solution and 100 μL of Reaction buffer additive for 500 μL. Sections are incubated for 45 minutes at room temperature with 100 μL of Click-iT EdU reaction buffer then washed with PBS and stain for fluorescent TRAP (ELF 97) and TO-PRO-3 (nuclear stain). Mounting media was 75% glycerol in PBS.
  • Microscopy. Images were acquired using an inverted Zeiss LSM880 laser scanning confocal microscope. Histological sections of 15 μm thickness were tile-scanned at 1.0 μm Z stacks.
  • Analysis. Sections were analyzed using Imaris in 3D view and individual Z stacks to quantify the percentage of EdU+ TRAP+ multinuclear cells and EdU+ nuclei per individual osteoclasts.
  • Adoptive transfers. Bone marrow monocytes were isolated from 12-16 weeks old donor. In fate-mapping experiments 1×106 total cells from Csf1riCre; Rosa26LSL-tdTomato mice were transferred at day 0, day 3 and day 6 by retro-orbital injections into recipient Csf1riCre; Rosa26LSL-YFP age and gender matched mice. Mice were sacrificed at day 11 and day 60 after the first transfer. For rescue of osteoclast activity 1×106 total bone marrow monocytes from Csf1riCre; Rosa26LSL-YFP or Csf1riCre; Rosa26LSL-tdTomato were transferred intra-peritoneal at p5, p8, and p11. Mice were sacrificed at p14 5 days after the last transfer. Bone marrow and blood was analyzed for the percentage of chimerism. To enrich Ly6C+ cells the Monocyte Isolation Kit (BM) for mouse (MACS Miltenyi Biotec, 130-100-629) was used as indicated by the manufacturer. Cell numbers of Ly6C+ cells were calculated by determining the cell number/ml using a Neubauer chamber in combination with a staining for Ly6C analyzed by flow cytometry. Bone samples were dissected and prepared for frozen sections as described above and stained for anti-GFP, anti-RFP, fluorescent TRAP and TO-PRO-3. The percentages of YFP+ or Tomato+ multinuclear cells was quantified in femurs of recipients.
  • Preparation and analysis of bone for histology of paraffin embedded samples. Bone samples were fixed in 4% formaldehyde (Fisher, 28908) in PBS for 1 day (embryo) or 3 days (post-natal mice) at 4° C. then washed 3 times with PBS and decalcified (for mice older than P7) in a 14% EDTA pH7.1 solution at 4° C. for 5 days to 15 days, washed 3 times with PBS and dehydrated in 70% ethanol for 1 day and processed for paraffin sections. Longitudinal sections of femurs were cut at 5 μm thickness using a Leica RM2265 paraffin microtome then place in Superfrost microscope slides let dry for 48 hours and heated in a dry incubator at 65° C. for 1 hour, dewaxed and stained for TRAP and hematoxylin.
  • Tartrate Resistant Acid Phosphatase (TRAP) staining protocol. To stain for TRAP, slides were placed in coplin jars and incubated in a 1% (v/v) Naphtol-Ether substrate solution in basic stock solution for 1 hr at 37° C. followed by incubation in a solution containing 2% (v/v) sodium nitrate solution and 2% (v/v) basic fuchsin solution in basic stock solution for 20 mins at 37° C. Slides are rinse in 3 changes of water then stained with hematoxylin solution (Sigma, GHS332) diluted 1:4 in water for 2 minute, washed 3 times with water then dehydrated and mounted in Entellan (Millipore, 107960). Solutions for TRAP staining. Basic stock solution: 0.92% (w/v) anhydrous sodium acetate (Sigma, 58750), 1.14% (w/v) dibasic dihydrate sodium tartrate (Sigma. S4797) and 0.28% (v/v) glacial acetic acid (Sigma, 537020) in distilled water, pH was adjusted between 4.7-5.0 with 5M sodium hydroxide (Fisher, S318-1). Napthol-Ether substrate solution: 2% (w/v) Napthol AS-BI. Phosphate (Sigma, 70482) in 2-Ethoxyethanol (Sigma, 256374). Sodium nitrate solution: 4% (w/v) Sodium nitrate (Sigma, 237213) in water. Basic fuchsin solution: 5% (w/v) basic fuchsin dye (Sigma, 857343) in 2N HCL (Fisher, A144-500).
  • Individual images from histological sections of 5 μm thickness were acquired using a Zeiss Axio Lab.A1 light microscope with a N-Achroplan 2.5X/0.07 M27 (420920-9901) or a N-Achroplan 20X/0.45 M27 (420950-9901) objective. Pictures were taken in ZEN lite software and exported as tiff files. Panoramic images were created with the photo-merge tool in Adobe Photoshop CS6. Pictures of mice were acquired with a dissecting microscope Leica M80 equipped with a Leica IC80 HD camera at 1.0× magnification. The region of interest analyzed was the methaphyseal trabecular bone 2 mm below the growth plate. The numbers of TRAP+ multinuclear cells (more than 3 nuclei per cell), associated to bone tissue, were quantified in ImageJ using the Cell Counter plugin. Numerical values were plotted using GraphPad Prism. For Static and dynamic histomorphometry. Young and aged mice were injected (i.p.) twice with 15 mg/kg body weight Calcein (Sigma) dissolved in 1.4% NaHCO3/ PBS 2 and 3 days apart, respectively. Mice were sacrificed 2 days after the last calcein injection. Femora and tibiae were fixed in 4% PBS-buffered paraformaldehyde and dehydrated in an ascending ethanol series. Subsequently, bones were embedded in methacrylate and cut into 7 μm sections to assess the fluorescent calcein labels. Unstained sections were analyzed using fluorescence microscopy to determine the mineralized surface/bone surface (MS/BS), the mineral apposition rate (MAR), and the bone formation rate/bone surface (BFR/BS). To determine numbers of osteoclasts, bones were decalcified for one week using Osteosoft (Merck), dehydrated, and embedded into paraffin. Tartrate-resistant acid phosphatase staining was used to assess the osteoclast surface per bone surface (Oc.SBS) and number of osteoclasts per bone surface (N.OcBS). Bone sections were analyzed using the Osteomeasure software (Osteometrics, USA) following international standards.
  • Preparation of and analysis of bones for immunofluorescence on frozen sections. Samples were prepared as above and after decalcification and washing were soaked in 30% sucrose in PBS at 4° C. for 1-2 days. Tissue samples were placed in disposable histology plastic molds and embedded in FSC22 Frozen Section Compound Clear (Leica, 3801480) and placed on a flat surface of dry ice to let freeze.
  • Immunofluorescence. Bones were cut at 15 μm thickness using a Cryostat Leica CM3050S with high profile microtome blades (Leica Surgipath DB80 HS) and cryofilm (Section Lab Inc.) and let to dry for 48 hours at 4° C. Before staining with antibodies, sections were let to equilibrate at room temperature for 30 minutes, rehydrated with PBS (Fisher, 14190) 3 times for 5 min at room temperature. Sections were incubated with blocking buffer containing 0.25% BSA (Fisher BP1600), 10% normal goat serum (Life Technologies, PCN 5000) and 0.3% triton (Sigma, T8787) in PBS for 1 hr at room temperature. Sections were washed 2 times with PBS for 5 minutes. Sections stained with anti-GFP biotin antibody were first incubated with Biotin/Streptavidin blocking kit (Vector laboratories, SP2002). Streptavidin blocking solution is prepared by adding 4 drops of streptavidin solution to 1 mL of PBS/0.25% BSA, samples were incubated for 15 min, then washed once with PBS for 5 min. Biotin blocking solution is prepared by adding 4 drops of biotin solution to 1 mL of PBS/0.25% BSA, samples were incubated for 15 min then washed once with PBS for 5 min. Primary and secondary antibodies used are listed in Table 2. Sections were also stained with fluorescent TRAP and nuclear stain.
  • TABLE 2
    Fate-mapping models: Flt3Cre; Rosa26LSL-YFP, Cfs1rCre;
    Rosa26LSL-YFP Csf1rMeriCreMer; Rosa26LSL-YFP and Tnfrsf11aCre; Rosa26LSL-YFP
    Catalog Staining Incubating
    Conjugation Company number Concentration Dilution Buffer conditions
    Primary Goat Biotin Abcam ab6658 1 mg/mL 1::50  PBS/0.25% 4° C.
    Antibody polyclonal BSA/0.3% overnight
    anti-GFP triton (15 hrs)
    Secondary Streptavidin AlexaFluor488 Thermo S-11223 1::100 PBS/0.25% 2 hrs at
    Antibody Fisher BSA room
    Scientific temperature
    Parabiosis
    Primary Goat Biotin Abcam ab6658 1 mg/mL 1::50  PBS/0.25% 4° C.
    Antibodies polyclonal Unconjugated ab62341 1::200 BSA/0.3% overnight
    anti GFP triton (15 hrs)
    Rabbit
    polyclonal
    anti-RFP
    Secondary Streptavidin AlexaFluor488 Thermo S-11223 1 mg/mL 1::100 PBS/0.25% 2 hrs at
    Antibodies Goat anti-rabbit AlexaFluor555 Fisher A21430 2 mg/mL 1::200 BSA room
    IgG Scientific temperature
    Bone marrow monocyte transfusions and transplants
    Primary Goat Biotin Abcam ab6658 1 mg/mL 1::50  PBS/0.25% 4° C.
    Antibodies polyclonal Unconjugated Rockland 600-401-379 1::200 BSA/0.3% overnight
    anti GFP triton (15 hrs)
    Rabbit
    polyclonal
    anti-RFP
    Secondary Streptavidin AlexaFluor488 Thermo S-11223 1 mg/mL 1::100 PBS/0.25% 2 hrs at
    Antibodies Goat anti-rabbit AlexaFluor555 Fisher A21430 2 mg/mL 1::200 BSA room
    IgG Scientific temperature
    EdU staining*
    Primary Rat anti-mouse Unconjugated Abcam ab6160 1 mg/mL 1::200 PBS/0.25% 4° C.
    Antibody Tubulin BSA/0.3% overnight
    triton (15 hrs)
    Secondary Goat anti-rat AlexaFluor555 Thermo A21434 2 mg/mL 1::200 PBS/0.25% 2 hrs at
    Antibody IgG Fisher BSA room
    Scientific temperature
    *After antibody staining sections were stained with the Click-it EdU Alexa Fluor 488 Imaging Assay kit, as described in methods.
  • Fluorescent TRAP staining. Sections were prepared for fluorescent TRAP by incubating with TRAP incubation solution (112 mM sodium acetate, 76 mM sodium tartate, and 11 mM sodium nitrite, pH 4.1-4.3) at room temperature for 10 minutes. Buffer was removed and incubated with ELF97 substrate (Molecular Probes E6589, 2 mM) at a concentration of 125 μM in TRAP incubation solution for 15 min under UV light and washed 2 times with PBS for 5 minutes. Nuclear stain used was TO-PRO-3 Iodide (Fisher T3605) 1:4000 in PBS for 5 minutes. Mounting media was 75% glycerol in PBS. Images were acquired using an inverted Zeiss LSM880 laser scanning confocal microscope with Argon-ion 488 nm, Diode 405-30 nm, DPSS 561-10 nm, HeNe 633 nm laser lines and Plan-Apochromat 40X/1.4 N.A. DIC (UV) VIS-IR oil objective. Histological sections of 15 μm thickness were tile-scanned at 1.5 μm Z-stacks in ZEN black and processed using ZEN lite. Sections were analyzed using Imaris in 3D view and individual Z stacks to quantify TRAP+ multinuclear cells and YFP and Tomato labeling. Pictures of the region of interest were generated in tiff format and analyzed by ImageJ software using ROI manager to calculate the mean fluorescence intensity (MFI) of individual osteoclasts. Numerical values were plotted using GraphPad Prism.
  • Animal imaging by computed tomography. NanoSPECT/CT: Mice were anesthetized under Isofluorane anesthesia and placed on an imaging table containing an animal bed equipped with a nosecone for gas inhalation and body temperature stabilization. For 3 week old mice a mouse bed was used and for mice 8 weeks and older a rat bed was used. Whole-body imaging of mice was acquired using a NanoSPECT/CT scanner (Mediso) for non-invasive and longitudinal monitoring of the 3D skeletal structure. Each CT scan averaged 15 minutes and was acquired with an exposure time of 1,000 ms and 240 projections set at a pitch of 1 degree. The tube energy of the X-ray was 55KVp and 145 μA. The in-plane voxel size was medium generating a voxel size of 147 μm3. Reconstructed images were analyzed using In Vivo Scope 2.0 (Bioscan, Inc.) software.
  • microCT. For tri-dimensional X-ray imaging by micro computed tomography, mice were sacrificed and bones placed in 70% ethanol until scanning. Bone microarchitecture was analyzed using the vivaCT40 (Scanco Medical, Switzerland). Entire femora or humeri were imaged at a resolution of 10.5 μm (1 slice) with an X-ray energy of 70 kVp, 114 mA, and an integration time of 200 ms. The machine was routinely calibrated using hydroxyapatite phantoms for density and geometry. Trabecular bone in femora or humeri from old mice was assessed in the metaphysis 20 slices below the growth plate using 150 slices. The trabecular region within the cortical bones (P21 mice) was determined in the femoral midshaft (100 slices up, 100 slices down). Pre-defined scripts from Scanco were used for the evaluation.
  • Preparation of tissues and staining for Flow cytometry. Yolk sac from E10.5 embryos was digested for 60 min at 37° C. in PBS containing 5% FCS, Collagenase Type 4 (Worthington, final 4.2 U/ml) and DNAseI (Sigma-Aldrich, final 100 μg/ml). The digestion reaction was stopped by incubation with 12.5 mM EDTA. Fetal liver was gently dissociated between the frosted ends of glass slides, and was then digested for 30 minutes using the same digestion enzyme mix as yolk sac. Blood was collected from anesthetized mice by retro-orbital venous sinus bleeding or cardiac puncture using a 1 mL syringe and a 26G needle rinsed with 100 mM EDTA (sigma E4884). The collected blood was lysed with 3 mL of red blood cell lysis buffer (155 mM NH4Cl sigma A9434, 10 mM NaHCO3 sigma S5761 and 0.1 mM EDTA sigma E4884) for 5 mins, washed with 10 mL of FACS buffer (PBS, 0.5% BSA and 2 mM EDTA) and centrifuged at 320 g for 7 minutes at 4° C. All mice were perfused with 10 ml of PBS after blood withdrawal. Bone marrow cells were harvested by crushing or flushing femurs and tibias using a syringe and a 26G needle with 10 mL of PBS/5% FCS or RPMI supplemented with 10% FBS. Bone marrow was dissociated by gently pipetting up and down with a 1 mL or 10 mL pipette. Spleens were gently dispersed between frosted slides and digested for 30 min at 37° C. in PBS with 5% FCS containing Collagenase Type 4 (Worthington) at a final concentration of 4.2 U/ml and 100 μg/ml DNAseI (Sigma-Aldrich). The reaction was stopped by incubation with 12.5 mM EDTA. Peritoneal cells were harvested by flushing the peritoneal cavity with 10 mL PBS/5% FCS heated to 37° C. Adult brain and liver were dissected, cut into small fragments, and incubated at 37° C. for 30 min in enzyme mix consisting of PBS with 1 mg/ml collagenase D (Sigma, 11088882001), 100 U/ml DNase I (Sigma, DN25), 2.4 mg/ml of dispase (Fisher, 17105-041) and 3% FBS (ThermoFisher Scientific 10438026) or PBS containing 4.2 U/ml Collagenase type 4 (Worthington), 100 μg/ml DNAseI, 2.4 mg/ml of Dispase (Gibco) and 3% FCS at 37° C. for 30 min. After enzyme digestion all tissues were further dissociated by mechanical disruption using 100 μm cell strainers (Falcon, 352360) and a 3 ml syringe plunger in 6 well plates containing 4 mL or 5 mL of cold FACS buffer or PBS/5% FCS. Single cell suspensions were transferred to 5 mL FACS tubes and pelleted by centrifugation at 320 g for 7 minutes at 4° C. The cell pellets were resuspended in FACS buffer containing purified anti-mouse CD16/32 antibody (1:100, Biolegend 101301), 5% normal mouse (Fisher, 015-000-120), 5% normal rat (Fisher 012-000-120) and 5% normal rabbit serum (Fisher, 011-000-120) and incubated for 10 minutes on ice or directly stained using blocking antibodies in the staining mixtures. Samples were immunostained with fluorochrome-conjugated antibodies for 30 minutes on ice, and analyzed by flow cytometry using an LSRFortessa or an LSR II (BD-Bioscience). Full list of antibodies for flow cytometry is provided in Table 3. Each sample was stained with Hoechst (ThermoFisher Scientific, Hoechst 33258, 1 μg/ml) or DAPI (Applichem, 1 μg/25 ml) moments prior to flow cytometry acquisition.
  • TABLE 3
    Antibody Clone Fluorochrome Company Dilution
    CD3 145-2C11 bio eBioscience 1/1000 (BM, Y, FL)
    CD3 145-2C11 BV711 BD 1/100 (BM, SP, LV, K, BR)
    Pharmingen 1/400 (E)
    CD3 145-2C11 APC-Cy7 BD 1/100 (BL)
    Pharmingen
    CD3 145-2C11 PE Biolegend 1/100 (BM)
    CD3 17A2 eF450 eBioscience 1/200 (BL)
    CD11b M1/70 bio eBioscience 1/1000 (BM, Y, FL)
    CD11b M1/70 PE-Cy7 eBioscience 1/200 (BL, BM, SP, LV, K, BR, LN, E)
    CD11b M1/70 BV711 Biolegend 1/200 (BM)
    CD19 1D3 bio eBioscience 1/500 (BM, Y, FL)
    CD19 1D3 BV711 BD 1/100 (BM, SP, LV, K, BR)
    Pharmingen 1/200 (BL)
    CD19 1D3 PE-Cy7 BD 1/400 (BL)
    Pharmingen
    B220 RA3-6B2 bio eBioscience 1/400 (BM, Y, FL)
    Gr1 RB6-8C5 bio eBioscience 1/1000 (BM, Y, FL)
    NK1.1 PK136 bio eBioscience 1/2000 (BM, Y, FL)
    NKp46 29A1.4 BV711 BD 1/100 (BM, SP, LV, K, BR)
    Pharmingen
    NKp46 29A1.4 AlexaFluor647 BD 1/200 (BL)
    Pharmingen
    Ter119 Ter119 bio eBioscience 1/500 (BM, Y, FL)
    Ter119 Ter119 BV711 BD 1/200 (BM)
    biosciences
    CD11b M1/70 PE BD 1/1600 (BM)
    Pharmingen
    CD11b M1/70 APC-eF780 eBioscience 1/600 (SP, Y, FL, LV, LN, BR, P)
    CD11c N418 APC eBioscience 1/150 (SP, BM, YS FL, LV, LN, BR, P)
    CD45 30-F11 PE eBioscience 1/150 (SP, BM, Y, FL, LV, LN, BR, P)
    CD45 30-F11 APC-eF780 eBioscience 1/200 (Y, FL, P)
    1/50 (BM, SP, LV, K)
    1/100 (BR, LN, E)
    CD45 30-F11 eF450 eBioscience 1/50 (BM)
    CD48 HM48-1 BV421 BD 1/400 (BM, Y, FL)
    Pharmingen
    CD48 HM48-1 APC Biolegend 1/100 (BM)
    CD115 AFS98 APC eBioscience 1/150 (SP, BM, Y, FL, LV, LN, BR, P)
    CD115 AFS98 APC Biolegend 1/100 (BM)
    CD115 AFS98 BV605 Biolegend 1/200 (BL, BM, SP, LV, K, BR)
    CD115 AFS98 PE eBioscience 1/100 (SP, BM, LV, LN, BR, P)
    CD117 2B8 PE-Cy7 eBioscience 1/3000 (Y, FL)
    CD117 2B8 APC eBioscience 1/400 (BM, Y, FL)
    CD117 2B8 APC-Cy7 Biolegend 1/100 (BM)
    CD117 2B8 BV605 BD 1/100 (BM)
    biosciences
    CD150 TC15-12F1 PE-Cy7 BioLegend 1/25 (Y, FL)
    1/100 (BM)
    F4/80 BM8 PE-Cy5 eBioscience 1/300 (SP, Y, FL, LV, LN, BR, P)
    F4/80 BM8 eF450 eBioscience 1/100 (BM, BR, LN, E)
    Gr-1 RB6-8C5 PECy5.5 eBioscience 1/3000 (SP, BM, Y, FL, LV, LN, BR, P)
    Gr-1 RB6-8C5 eF450 eBioscience 1/1000 (Y, FL)
    Gr-1 RB6-8C5 BV711 Biolegend 1/100 (BM)
    Ly6C HK1.4 PerCpCy5.5 eBioscience 1/800 (Y, FL)
    Ly6C HK1.4 BV421 Biolegend 1/200 (BL)
    1/400 (SP, LV, K, BR)
    Ly6C HK1.4 BV510 Biolegend 1/200 (BM)
    Ly6C HK1.4 AlexaFluor488 Biolegend 1/50 (BM)
    Ly6G 1A8 BV510 Biolegend 1/100 (BM)
    1/200 (Bl, LN)
    1/400 (SP, LV, K, BR)
    Ly6G 1A8 BV711 BD 1/200 (BM)
    Pharmingen
    MHCII M5/114.15.2 Alexa Fluor 700 eBioscience 1/150 (SP, BM, Y, FL, LV, LN, BR, P)
    MHCII M5/114.15.2 AlexaFluor700 Biolegend 1/100 (BM)
    1/200 (BL, SP, LV, K, BR)
    Sca1 D7 PercpCy5.5 eBioscience 1/500 (BM, Y, FL)
    Sca-1 D7 BV421 BD 1/200 (BM)
    Pharmingen
    Sca-1 D7 BV711 Biolegend 1/100 (BM)
    Streptavidin V500 BD 1/800 (BM, Y, FL)
    Biosciences
    SiglecF E50-2440 AlexaFluor647 BD 1/100 (BM, LN)
    Pharmingen
    BM: Bone marrow; SP: Spleen; BL: Blood; Y: Yolk sac; FL: Fetal Liver; LV: Liver; LN: Lung; BR: Brain; P: PEC: E: Epidermis; K: Kidney.
  • All analysis was conducted using FlowJo (Tree Star). In all tissues single live cells were gated by exclusion of dead cells labeled positive by Hoechst or DAPI, side scatter (SSC-A) and forward scatter (FSC-A) and doublet exclusion using forward scatter width (FSC-W) against (FSC-A), as previously described. In order to calculate cell numbers per organ or per gram of tissue, organs were weighted, cell suspensions were prepared from a weighted amount (20 to 500 mg) of tissue, and the number of cells per gram of tissue was determined using a cell counter (GUAVA easyCyte HT).
  • Statistical analysis and reproducibility. Data are shown as mean with individual values per mouse represented as circles, unless stated otherwise. Statistical significance was analyzed with GraphPad Prism using unpaired t-tests and two-way ANOVA with Tukey's multiple comparisons test as indicated in the figure legends. Significance was considered at P value (P) *P<0.05; **P<0.01; ***P<0.001; ****P<0.0001. Then value represents biological replicates. Experiments were repeated to ensure reproducibility of the observations. Equal variance was assumed for cell-counting experiments. No statistical methods were used to predetermine sample size.
  • Example 1: Osteoclast Origins and Maintenance
  • In vitro, osteoclasts arise by fusion of HSC-derived precursors and require expression of Csf1r and Tnfrsf11a (Rank). To probe the origin of osteoclasts in vivo, Csf1Cre; Csf1rfl/fl; and Csf1rCre; Tnfrsf11afl/fl mice were generated. These mice presented with an osteopetrotic phenotype similar to Csf1 (op/op), Csf1r, and Tnfrsf11a mutants and characterized in young mice by lack of teeth eruption, skull and skeletal deformities with shortness of long bones, increased bone density, and lack of osteoclasts and hematopoietic cells (FIG. 1A; FIG. 5B). To confirm that osteoclast differentiation requires expression of Tnfrsf11a and Csf1r in HSC-derived precursors, Flt3Cre; Tnfrsf11afl/fl mice, Flt3Cre; Csf1rfl/fl mice, and VavCre; Csf1rfl/fl mice were generated. Surprisingly, all young mice had normal teeth (FIG. 1B) and bone morphology (FIG. 6A-C), normal bone marrow cellularity (FIG. 1C), and normal osteoclast numbers (FIG. 1D) in comparison to control littermates. However, Flt3Cre; Tnfrsf11afl/fl mice, Flt3Cre; Csf1rfl/fl mice, and VavCre; Csf1rfl/fl mice lost their osteoclasts over time (FIG. 1D), and by 22-60 weeks of age had increased trabecular bone density (FIG. 1E), decreased hematopoietic cell numbers in the long bones (FIG. 1C; FIG. 6D-G) and tri-dimensional X-ray imaging by micro computed tomography (micro-CT) confirmed increased bone mass while bone formation measured by calcein incorporation was similar to control (FIG. 1F, FIG. 7A-E).
  • Mice lacking Csf1r expression in Flt3Cre-expressing progenitors presented with a similar phenotype: Flt3Cre; Csf1rfl/fl mice have normal teeth and bone morphology at 4 week of age (FIG. 1B, FIG. 6C), but osteoclast number are decreased over time and bone mass is increased at 22 weeks of age (FIG. 1D-E, FIG. 7A-E). In a complementary approach to delete Csf1r in HSC-derived progenitors, VavCre; Csf1rfl/fl mice were generated. Again, VavCre; Csf1rfl/fl mice were not osteopetrotic at birth, had normal teeth (FIG. 1B), and hematopoietic cell numbers in long bones at 4 week of age (FIG. 1C). However, they developed a late-onset osteopetrosis similar to Flt3rCre; Tnfrsf11afl/fl and Flt3Cre; Csf1rfl/fl mice: at ˜60 weeks of age VavCre; Csf1rfl/fl mice had decreased osteoclasts and hematopoietic cell numbers in long bones (FIG. 1C-D), enlarged trabecular bone (FIG. 1E) and increased bone mass (FIG. 1F, FIG. 7A-E). These data suggested that osteoclast development, tooth eruption and the development of bone and the bone marrow cavity requires precursors that express Csf1r, but not Flt3Cre or VavCre although postnatal contribution of Flt3Cre/VavCre expressing cells is important for optimal osteoclast function in adults and aging mice.
  • Fetal expression of Csf1rCre and Flt3Cre was analyzed using a fluorescent reporter in Csf1rCre; Rosa26LSL-YFP and Flt3Cre; Rosa26LSL-YFP mice (FIG. 1 G-I). TRAP+ multinucleated cells that appear at embryonic day (E)15 in ossification centers are labeled with YFP in Csf1rCre; Rosa26LSL-YFP mice and osteoclasts remain YFP positive throughout life (FIG. 1 G,I; FIG. 8A-E), but gain expression of YFP after birth in Flt3Cre; Rosa26LSL-YFP mice despite colonization of the fetal bone marrow by Flt3Cre+ YFP+ hematopoietic cells (FIG. 1 H; FIG. 8A-E). These data suggested that although postnatal contribution of HSC-derived cells is important for optimal osteoclast maintenance and function in adults and aging mice, osteoclast development, tooth eruption, the development of bone, and the bone marrow cavity requires precursors independent from the HSC lineage, possibly from the embryonic EMP lineage of resident macrophages because Csf1rCre mice allow deletion of target genes in both the embryonic EMP lineage and in the HSC lineage, while Flt3Cre and VavCre (FIG. 9A-D) are not expressed in the EMP lineage.
  • In support of this hypothesis, it was found that TRAP+ multinucleated cells develop in ˜E15 ossification centers from Myb-deficient embryo, which lack HSC but still support the development of EMP-derived macrophages (FIG. 2A, B). In addition, TRAP+ multinucleated cells are labeled with YFP in Csf1rMer-iCre-Mer; Rosa26LSL-YFP mice pulsed at E8.5 with a single dose of hydroxytamoxifen (4-OHT), which labels EMP but not HSC (FIG. 2C, D; FIG. 8A-E). Altogether, these results indicate that fetal osteoclasts arise from EMP in ossification centers.
  • Whether EMP are required for bone development was investigated. Tnfrsf11a is expressed by osteoclasts, but its expression is also a hallmark of EMP-derived pMacs that colonize the developing embryo. In two independent lines of Tnfrsf11aCre knock-in mice, Cre-mediated expression of a Rosa26LSL-YFP fluorescent reporter is achieved with high efficiency in fetal macrophages but with low efficiency or not at all in HSC and their progeny in blood and tissues (FIG. 9A-D). It was therefore hypothesized that conditional deletion of Csf1r in Tnfrsf11aCre would recapitulate macrophage deficiency observed in Csf1r-deficient mice, while leaving the HSC lineage unaffected. To test osteoclast and bone development in this model, two independent lines of Tnfrsf11aCre; Csf1rfl/fl mice were generated (FIG. 2E-J). Tnfrsf11aCre; Csf1rfl/fl lack tissue macrophages such as brain microglia and epidermal Langerhans cells at birth while development of HSCs and blood cells was preserved (FIG. 6A-N). They present with a severe osteopetrotic phenotype including the lack of tooth eruption (FIG. 2E), misshaped skulls and short long bones (FIG. 2F-H; FIG. 5A-G) with few osteoclasts (FIG. 2I), increased bone density and initially lack a bone marrow cavity (FIG. 2F; FIG. 6A-N). In contrast to Csf1r-deficient mice, however, osteoclasts and hematopoietic cells progressively colonized the long bones of Tnfrsf11aCre; Csf1rfl/fl mice during the first month of life (FIG. 2I; FIG. 5A-G; FIG. 6A-N) although the mice remained toothless and skull and long bone deformity persisted (FIG. 2E-H; FIG. 5A-G). Calcein incorporation was similar in Tnfrs11aCre; Csf1rfl/fl mice and their littermate controls (FIG. 10A-F). In a complementary approach ablation of Csf1r expression in E10.5 embryos using a single dose of tamoxifen in Rosa26-creERT2+; Csf1rfl/− mice resulted in defective tooth eruption in 3 out of 4 pups at 21 days of age (FIG. 9A-D). Altogether, this analysis supports a model in which EMP-derived embryonic osteoclasts are needed for teeth eruption, normal skull shape and optimal formation of long bones, and the timely colonization of long bones by hematopoietic progenitors, while HSC-derived osteoclasts are important for the maintenance of bone mass after birth and later in life, although they may partially rescue bone development in the absence of EMP-derived osteoclasts in Tnfrs11aCre; Csf1rfl/fl mice.
  • Example 2: Therapeutic Approach for the Treatment of Osteopetrosis
  • To probe the mechanisms that underlie the contribution of HSC-derived blood cells to osteoclast maintenance as well as the lifespan and dynamics of osteoclasts in vivo, time-course parabiosis experiments were performed (FIG. 3A). After 4 to 8 weeks of shared blood circulation between Csf1rCre; Rosa26LSL-YFP and Csf1rCre; Rosa26LSL-tdTomato parabionts, all osteoclasts, defined as TRAP+ multinucleated cells lining the bone surface, expressed both YFP and tdTomato within the same cells (FIG. 3A, B). No other cell was found to co-express YFP and tdTomato in bones (FIG. 3B). This is compatible with the presence of nuclei from both partners in individual osteoclasts. Moreover, when parabionts were separated after 4 weeks (FIG. 3C), most recipient YFP+ osteoclasts retained tdTomato staining 14 weeks after separation, and two third of osteoclasts from former parabionts still expressed both YFP and tdTomato 24 weeks after separation (FIG. 3C). TdTomato signal intensity per YFP osteoclast increased during the 8-week period of shared blood circulation, and decreased after separation (FIG. 11A-C). Most murine osteoclasts have ˜5 (3 to 7) nuclei, with a modest increase of nuclei per cell between 1 month and 6 months of age (FIG. 3E). These data therefore suggest that individual osteoclast syncytia are long lived, but acquire new nuclei one by one every 4 to 8 weeks from circulating blood cells so that it takes more than 6 months to renew all 5 nuclei from a given osteoclast.
  • The number and fusion-rate of HSC-derived nuclei acquired by osteoclasts in short-term 5-ethynyl-2′-deoxyuridine (EdU) incorporation studies was calculated. A single intravenous pulse of EdU (20 μg/g) labels mitotic nuclei, is bioavailable in the bone marrow for ˜90 min, and ˜50% of bone marrow and blood monocytic cells are EdU+ for ˜48 hrs (FIG. 11A-C). It was observed that ˜1 to 2% of osteoclasts were labelled after 72 hrs, with only 1 EdU+ nucleus per osteoclast in 90% of positive cells (FIG. 3F) suggesting acquisition by osteoclasts of single post-mitotic nuclei at a time. The proportion of osteoclasts acquiring a new nucleus can be estimated ˜0.5 to 2% per day in this model, compatible with individual nuclei being replaced every˜2 months. Altogether the studies above suggested a model (FIG. 3G) where: osteoclasts that control skeletal development develop in ossification centers from EPM, while their post-natal maintenance is mediated via the serial acquisition by long-lived syncytia of new nuclei from HSC-derived blood leukocytes, rather than by de novo renewal by lateral fusion or proliferation of osteoclast precursors.
  • A prediction from this model is that osteopetrosis due to a recessive mutation affecting osteoclasts function may be rescued or prevented through parabiosis with a wild-type partner. Parabiosis experiments between 4 week-old cathepsin K deficient mice, which develop an adult-onset form of osteopetrosis known as pycnodysostosis, and cathepsin K+/− or cathepsin K−/− littermates and between wild type mice as control showed a reduction of bone volume in 10 weeks old cathepsin K−/− mice paired with cathepsin K+/− littermates (FIG. 4A), suggesting that circulating blood cells carrying a wild type cathepsin K allele are sufficient to reduce bone density. To confirm that expression of a donor-derived gene by recipient osteoclasts results from fusion with blood circulating monocytic cells, intravenous injections of Kit Ly6C+ cells from the bone marrow of Csf1rCre; Rosa26LSL-tdTomato into Csf1rCre; Rosa26LSL-YFP recipients were performed (FIG. 4B; FIG. 12A-C). This resulted in stable expression of tdTomato in 20-40% of osteoclasts 1 week and 8 weeks after transfusion, in the absence of other donor-derived blood cells or bone marrow progenitors (FIG. 4B, C; FIG. 12A-C). These results suggest that parabiosis or an appropriate regimen of transfusion can achieve expression of a donor-derived gene by recipient osteoclasts in the absence of HSC chimerism, and in addition that this effect can last for several months.
  • Partial rescue of osteopetrosis occurs postnatally in Tnfrsf11aCre; Csf1rfl/fl mice, suggesting that transfusion of monocytic cells may also be able to rescue bone development in early-onset congenital osteopetrosis in the absence of a bone marrow transplantation. Intra-peritoneal injections of Kit Ly6C+ monocytic cells from Csf1rCre; Rosa26LSL-YFP into Csf1rCre; Csf1rF/F neonates starting from post-natal day 5, resulted in complete or partial rescue of teeth eruption (FIG. 4D) and long bone development as assessed by CT-Scan, with the development of a bone marrow cavity (FIG. 4D) at day 14 in infant mice from 3 different litters (FIG. 4D, FIG. 13). In all mice, histology of femurs indicated the presence of numerous YFP+ TRAP+ osteoclasts lining the bone (FIG. 4E, F). Histology and flow cytometry analyses showed the lack of YFP+ circulating blood cells or bone marrow progenitors (FIG. 4E, G), indicating absence of HSC engraftment. These data suggest that transfusion of monocytic cells can rescue bone development in early-onset autosomal recessive osteopetrosis in infant mice, in the absence of HSC transplantation.
  • Summary
  • The results described above show that osteoclasts originating from EMP are essential for normal bone development. Moreover, it is shown that osteoclasts are long-lived in adult and their function is maintained by iterative fusion of individual HSC-derived circulating cells with existing syncytia. In the absence or deficiency of EMP-derived osteoclasts, however, their timely replacement by transfusion with monocytic cells can rescue bone development in early-onset osteopetrotic mice in the absence of bone marrow transplantation. This could contribute to the treatment of early onset osteopetrosis, since the current treatment by bone marrow transplantation. This is of potential clinical relevance because bone marrow/HSC transplantation, the standard of care in early-onset osteopetrosis in mice and human, requires irradiation or chemotherapy which carry the risk of threatening infections and is frequently performed late in patients who already suffer severe complications and is plagued by a ˜48% overall survival at 6-years. In addition, the original mechanism that mediates osteoclast maintenance suggests that they represent a unique target for gene transfer by cellular therapies based on transfusion of wild type or engineered monocytic cells to modulate osteoclast activity and bone remodeling in adults. Accordingly, these results demonstrate that monocytic cells may be effective in methods for treating osteopetrosis, and to modulate osteoclast activity and bone remodeling.
  • Example 3: Ex Vivo Gene Therapy for the Treatment of a Form of Osteopetrosis: Pycnodysostosis
  • This example demonstrates the prophetic use of monocytic cells obtained from a subject and engineered to express a gene, such as CTSK, for the treatment of osteopetrosis in the subject.
  • Pycnodysostosis is an autosomal recessive form of osteopetrosis, due to loss of function mutations in the cathepsin K gene (CTSK), compatible with life, and characterized by short stature, deformity of the skull maxilla and phalanges, increased density of the bones, osteosclerosis, and fragility of bone. There is no treatment for this disease as the relatively comparably milder symptoms do not qualify for bone marrow transplantation.
  • Subjects suspected of having or diagnosed as having cathepsin K deficiency will be selected for a gene therapy trial and will undergo GMP grade collection of peripheral blood monocytes by apheresis and/or elutriation. Purified monocytes will then be transduced with a lentivirus designed to express a cDNA coding for the wild type allele of CTSK under a strong promoter (e.g., CMV). Transduced monocytes will then be washed to remove contaminating viral particles and rested for 12 hours followed by reinfusion to the patient. Assuming ˜109 monocytes can be collected, the number of cells transferred per kilogram of body weight would be >10′. Following the auto-transfusion, the subject will be monitored at regular intervals for clinical and radiological signs of pycnodysostosis in order to determine the efficiency of the procedure and the frequency with which it should be repeated.
  • It is predicted that subjects suspected of having or diagnosed as having pycnodysostosis and receiving therapeutically effective amounts of monocytic cells engineered to express CTSK will display reduced severity or elimination of one or more symptoms associated with pycnodysostosis. Accordingly, these results will show that transfusion of monocytic cells engineered to express the wild-type version of one or more genes implicated in osteopetrosis is useful in methods for treating subjects in need thereof for the treatment of osteopetrosis.
  • Example 4: Ex Vivo Gene Therapy for the Treatment of a Form of Osteopetrosis: Severe Infantile Autosomal Recessive Osteopetrosis
  • This example demonstrates the prophetic use of monocytic cells obtained from a subject and engineered to express a gene, such as TCIRG1, TNFRSF11a, CA2, CLCN7, OSTM1, and/or PLEKHM1, for the treatment of osteopetrosis in the subject.
  • Infants diagnosed with severe infantile autosomal recessive osteopetrosis with defects in TCIRG1, TNFRSF11a, CA2, CLCN7, OSTM1, and/or PLEKHM1 will be eligible for an early auto-transfusion of corrected monocytes (as described in Example 3), to try to prevent early developmental complications, including blindness and skeletal deformities, and increase the success of bone marrow transplantation, by restoring a bone marrow niche. The protocol will be carried out in a manner similar to that of Example 3, and monocytes will be transduced with a lentivirus designed to express a cDNA coding for the wild type allele of the deficient gene under a strong promoter, with a similar number of transferred cells per kg of weight. Following the auto-transfusion, subjects will be monitored at regular intervals for clinical and radiological signs, and possibly bone biopsy in order to determine the efficiency of the procedure. In addition, collection of monocytes and hematopoietic stem cells/progenitors at the same time would allow to proceed with the auto transfusion of transduced monocytes, while stem cells would undergo gene editing in view of auto transplantation and long term genetic rescue.
  • It is predicted that subjects suspected of having or diagnosed as having severe infantile autosomal recessive osteopetrosis and receiving therapeutically effective amounts of monocytic cells engineered to express TCIRG1, TNFRSF11a, CA2, CLCN7, OSTM1, and/or PLEKHM1 will display reduced severity or elimination of one or more symptoms associated with severe infantile autosomal recessive osteopetrosis. Accordingly, these results will show that transfusion of monocytic cells engineered to express the wild-type version of one or more genes implicated in osteopetrosis is useful in methods for treating subjects in need thereof for the treatment of osteopetrosis.
  • REFERENCES
  • The following references are incorporated by reference.
    • 1. Coccia, P. F. et al. Successful bone-marrow transplantation for infantile malignant osteopetrosis. The New England journal of medicine 302, 701-708, doi:10.1056/NEJM198003273021301 (1980).
    • 2. Sorell, M. et al. Marrow transplantation for juvenile osteopetrosis. The American journal of medicine 70, 1280-1287 (1981).
    • 3. Lacey, D. L. et al. Osteoprotegerin ligand is a cytokine that regulates osteoclast differentiation and activation. Cell 93, 165-176 (1998).
    • 4. Wiktor-Jedrzejczak, W. et al. Total absence of colony-stimulating factor 1 in the macrophage-deficient osteopetrotic (op/op) mouse. Proceedings of the National Academy of Sciences 87, 4828-4832, doi:10.1073/pnas.87.12.4828 (1990).
    • 5. Kong, Y. Y. et al. OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis. Nature 397, 315-323, doi:10.1038/16852 (1999).
    • 6. Dougall, W. C. et al. RANK is essential for osteoclast and lymph node development. Genes & development 13, 2412-2424 (1999).
    • 7. Hsu, H. et al. Tumor necrosis factor receptor family member RANK mediates osteoclast differentiation and activation induced by osteoprotegerin ligand. Proceedings of the National Academy of Sciences of the United States of America 96, 3540-3545 (1999).
    • 8. Yoshida, H. et al. The murine mutation osteopetrosis is in the coding region of the macrophage colony stimulating factor gene. Nature 345, 442-444 (1990).
    • 9. Dai, X.-M. et al. Targeted disruption of the mouse colony-stimulating factor 1 receptor gene results in osteopetrosis, mononuclear phagocyte deficiency, increased primitive progenitor cell frequencies, and reproductive defects. Blood 99, 111-120, doi:10.1182/blood.V99.1.111 (2002).
    • 10. Luchin, A. et al. Genetic and physical interactions between Microphthalmia transcription factor and PU.1 are necessary for osteoclast gene expression and differentiation. The Journal of biological chemistry 276, 36703-36710, doi:10.1074/jbc.M106418200 (2001).
    • 11. Grigoriadis, A. E. et al. c-Fos: a key regulator of osteoclast-macrophage lineage determination and bone remodeling. Science 266, 443-448 (1994).
    • 12. Lomaga, M. A. et al. TRAF6 deficiency results in osteopetrosis and defective interleukin-1, CD40, and LPS signaling. Genes & development 13, 1015-1024 (1999).
    • 13. Tondravi, M. M. et al. Osteopetrosis in mice lacking haematopoietic transcription factor PU.1. Nature 386, 81-84, doi:10.1038/386081a0 (1997).
    • 14. Takayanagi, H. et al. Induction and activation of the transcription factor NFATc1 (NFAT2) integrate RANKL signaling in terminal differentiation of osteoclasts. Developmental cell 3, 889-901 (2002).
    • 15. Lau, R. Y. & Guo, X. A review on current osteoporosis research: with special focus on disuse bone loss. J Osteoporos 2011, 293808, doi:10.4061/2011/293808 (2011).
    • 16. Walker, D. G. Bone resorption restored in osteopetrotic mice by transplants of normal bone marrow and spleen cells. Science 190, 784-785 (1975).
    • 17. Walker, D. G. Control of bone resorption by hematopoietic tissue. The induction and reversal of congenital osteopetrosis in mice through use of bone marrow and splenic transplants. The Journal of experimental medicine 142, 651-663 (1975).
    • 18. Ballet, J. J., Griscelli, C., Coutris, C., Milhaud, G. & Maroteaux, P. Bone-marrow transplantation in osteopetrosis. Lancet 2, 1137 (1977).
    • 19. Nisbet, N. W., Menage, J. & Loutit, J. F. Bone-marrow transplantation in osteopetrosis. Lancet 2, 1236 (1977).
    • 20. Ash, P., Loutit, J. F. & Townsend, K. M. Osteoclasts derived from haematopoietic stem cells. Nature 283, 669-670 (1980).
    • 21. Orchard, P. J. et al. Hematopoietic stem cell transplantation for infantile osteopetrosis. Blood 126, 270-276, doi:10.1182/blood-2015-01-625541 (2015).
    • 22. Xiao, Y. et al. Identification of the Common Origins of Osteoclasts, Macrophages, and Dendritic Cells in Human Hematopoiesis. Stem cell reports 4, 984-994, doi:10.1016/j.stemcr.2015.04.012 (2015).
    • 23. Shalhoub, V. et al. Characterization of osteoclast precursors in human blood. British journal of haematology 111, 501-512 (2000).
    • 24. Stadtfeld, M. & Graf, T. Assessing the role of hematopoietic plasticity for endothelial and hepatocyte development by non-invasive lineage tracing. Development 132, 203-213, doi:10.1242/dev.01558 (2005).
    • 25. Taniguchi, N. et al. Stage-specific secretion of HMGB 1 in cartilage regulates endochondral ossification. Molecular and cellular biology 27, 5650-5663, doi:10.1128/MCB.00130-07 (2007).
    • 26. Schulz, C. et al. A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science 336, 86-90, doi:10.1126/science. 1219179 (2012).
    • 27. Gomez Perdiguero, E. et al. Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors. Nature 518, 547-551, doi:10.1038/nature13989 (2015).
    • 28. Mass, E. et al. Specification of tissue-resident macrophages during organogenesis. Science 353, doi:10.1126/science.aaf4238 (2016).
    • 29. Deng, L. et al. A novel mouse model of inflammatory bowel disease links mammalian target of rapamycin-dependent hyperproliferation of colonic epithelium to inflammation-associated tumorigenesis. Am J Pathol 176, 952-967, doi:10.2353/ajpath.2010.090622 (2010).
    • 30. Qian, B. Z. et al. CCL2 recruits inflammatory monocytes to facilitate breast-tumour metastasis. Nature 475, 222-225, doi:10.1038/nature10138 (2011).
    • 31. Li, J., Chen, K., Zhu, L. & Pollard, J. W. Conditional deletion of the colony stimulating factor-1 receptor (c-fms proto-oncogene) in mice. Genesis 44, 328-335, doi:10.1002/dvg.20219 (2006).
    • 32. Dai, X. M. et al. Targeted disruption of the mouse colony-stimulating factor 1 receptor gene results in osteopetrosis, mononuclear phagocyte deficiency, increased primitive progenitor cell frequencies, and reproductive defects. Blood 99, 111-120 (2002).
    • 33. Benz, C., Martins, V. C., Radtke, F. & Bleul, C. C. The stream of precursors that colonizes the thymus proceeds selectively through the early T lineage precursor stage of T cell development. The Journal of experimental medicine 205, 1187-1199, doi:10.1084/jem.20072168 (2008).
    • 34. Mucenski, M. L. et al. A functional c-myb gene is required for normal murine fetal hepatic hematopoiesis. Cell 65, 677-689 (1991).
    • 35. Stadtfeld, M. & Graf, T. Assessing the role of hematopoietic plasticity for endothelial and hepatocyte development by non-invasive lineage tracing. Development 132, 203-213, doi:10.1242/dev.01558 (2005).
    • 36. Hanada, R. et al. Central control of fever and female body temperature by RANKL/RANK. Nature 462, 505-509, doi:10.1038/nature08596 (2009).
    • 37. Maeda, K. et al. Wnt5a-Ror2 signaling between osteoblast-lineage cells and osteoclast precursors enhances osteoclastogenesis. Nature medicine 18, 405-412, doi:10.1038/nm.2653 (2012).
    • 38. Hameyer, D. et al. Toxicity of ligand-dependent Cre recombinases and generation of a conditional Cre deleter mouse allowing mosaic recombination in peripheral tissues. Physiol Genomics 31, 32-41, doi:10.1152/physiolgenomics.00019.2007 (2007).
    • 39. Srinivas, S. et al. Cre reporter strains produced by targeted insertion of EYFP and ECFP into the ROSA26 locus. BMC developmental biology 1, 4 (2001).
    • 40. Madisen, L. et al. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nat Neurosci 13, 133-140, doi:10.1038/nn.2467 (2010).
    • 41. Schulz, C. et al. A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science 336, 86-90, doi:10.1126/science. 1219179 (2012).
    • 42. Gomez Perdiguero, E. et al. Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors. Nature 518, 547-551, doi:10.1038/nature13989 (2015).
    • 43. Kamran, P. et al. Parabiosis in mice: a detailed protocol. J Vis Exp, doi:10.3791/50556 (2013).
    • 44. Jiang, X. et al. Histological analysis of GFP expression in murine bone. J Histochem Cytochem 53, 593-602, doi:10.1369/jhc.4A6401.2005 (2005).
    • 45. Filgueira, L. Fluorescence-based staining for tartrate-resistant acidic phosphatase (TRAP) in osteoclasts combined with other fluorescent dyes and protocols. J Histochem Cytochem 52, 411-414, doi:10.1177/002215540405200312 (2004).
    • 46. Mass, E. et al. Specification of tissue-resident macrophages during organogenesis. Science 353, doi:10.1126/science.aaf4238 (2016).
    • 47. Arndt, K. et al. SETD1A protects HSCs from activation-induced functional decline in vivo. Blood 131, 1311-1324, doi:10.1182/blood-2017-09-806844 (2018).
    • 48. Grinenko, T. et al. Clonal expansion capacity defines two consecutive developmental stages of long-term hematopoietic stem cells. The Journal of experimental medicine 211, 209-215, doi:10.1084/jem.20131115 (2014).
  • The present technology is not to be limited in terms of the particular embodiments described in this application, which are intended as single illustrations of individual aspects of the present technology. Many modifications and variations of this present technology can be made without departing from its spirit and scope, as will be apparent to those skilled in the art. Functionally equivalent methods and apparatuses within the scope of the present technology, in addition to those enumerated herein, will be apparent to those skilled in the art from the foregoing descriptions. Such modifications and variations are intended to fall within the scope of the appended claims. The present technology is to be limited only by the terms of the appended claims, along with the full scope of equivalents to which such claims are entitled. It is to be understood that this present technology is not limited to particular methods, reagents, compounds compositions or biological systems, which can, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting.
  • In addition, where features or aspects of the disclosure are described in terms of Markush groups, those skilled in the art will recognize that the disclosure is also thereby described in terms of any individual member or subgroup of members of the Markush group.
  • As will be understood by one skilled in the art, for any and all purposes, particularly in terms of providing a written description, all ranges disclosed herein also encompass any and all possible subranges and combinations of subranges thereof. Any listed range can be easily recognized as sufficiently describing and enabling the same range being broken down into at least equal halves, thirds, quarters, fifths, tenths, etc. As a nonlimiting example, each range discussed herein can be readily broken down into a lower third, middle third and upper third, etc. As will also be understood by one skilled in the art all language such as “up to,” “at least,” “greater than,” “less than,” and the like, include the number recited and refer to ranges which can be subsequently broken down into subranges as discussed above. Finally, as will be understood by one skilled in the art, a range includes each individual member. Thus, for example, a group having 1-3 cells refers to groups having 1, 2, or 3 cells. Similarly, a group having 1-5 cells refers to groups having 1, 2, 3, 4, or 5 cells, and so forth.
  • All patents, patent applications, provisional applications, and publications referred to or cited herein are incorporated by reference in their entirety, including all figures and tables, to the extent they are not inconsistent with the explicit teachings of this specification.
  • Other embodiments are set forth within the following claims.

Claims (21)

What is claimed is:
1. A method for treating or preventing osteopetrosis in a subject in need thereof, the method comprising administering a composition comprising a therapeutically effective amount of monocytic cells from a healthy donor to the subject.
2. The method of claim 1, wherein the subject is characterized by decreased expression of one or more of CA2, CLCN7, CTSK, CSF1R, IKBKG, ITGB3, OSTM1, PLEKHM1, TCIRG1, TNFRSF11A, and TNFSF11, as compared to the monocytic cells of the donor.
3. The method of claim 1, wherein the osteopetrosis comprises one or more of stunted growth, skeletal deformity, increased likelihood of bone fracture, anemia, recurrent infections, hepatosplenomegaly, facial paralysis, abnormal cortical bone morphology, abnormal form of vertebral bodies, abnormal temperature regulation, abnormality of the ribs, abnormality of vertebral epiphysis morphology, bone pain, cranial nerve paralysis, craniosynostosis, hearing impairment, and hypocalcemia.
4. The method of claim 1, wherein the composition is formulated for intravenous administration by injection, infusion, or transfusion.
5. The method of claim 1, wherein the subject is a mammal.
6. The method of claim 5, wherein the mammalian subject is a human.
7. The method of claim 1, wherein the subject is characterized by a cathepsin K deficiency.
8. A method for treating or preventing osteopetrosis in a subject in need thereof, the method comprising administering to the subject a composition comprising a therapeutically effective amount of monocytic cells engineered to express one or more genes selected from CA2, CLCN7, CTSK, CSF1R, IKBKG, ITGB3, OSTM1, PLEKHM1, TCIRG1, TNFRSF11A, and TNFSF11.
9. The method of claim 7, wherein the osteopetrosis comprises one or more of stunted growth, skeletal deformity, increased likelihood of bone fracture, anemia, recurrent infections, hepatosplenomegaly, facial paralysis, abnormal cortical bone morphology, abnormal form of vertebral bodies, abnormal temperature regulation, abnormality of the ribs, abnormality of vertebral epiphysis morphology, bone pain, cranial nerve paralysis, craniosynostosis, hearing impairment, and hypocalcemia.
10. The method of claim 8, wherein the composition is formulated for intravenous administration by injection, infusion, or transfusion.
11. The method of claim 8, wherein the subject is a mammal.
12. The method of claim 11, wherein the mammalian subject is a human.
13. The method of claim 8, wherein the subject is characterized by a cathepsin K deficiency.
14. The method of claim 8, wherein the monocytic cells are obtained from the subject.
15. A donor monocytic cell line engineered to express one or more genes selected from CA2, CLCN7, CTSK, CSF1R, IKBKG, ITGB3, OSTM1, PLEKHM1, TCIRG1, TNFRSF11A, and TNFSF11, wherein the one or more genes is operably linked to a heterologous nucleic acid to form a chimeric nucleic acid construct.
16. The donor monocytic cell line of claim 15, wherein the heterologous nucleic acid encodes a selectable marker.
17. The donor monocytic cell line of claim 16, wherein the selectable marker is a bioluminescent protein, a fluorescent protein, a chemiluminescent protein, a xanthine-guanine phosphoribosyl transferase gene (gpt), or any combination thereof.
18. The donor monocytic cell line of claim 15, wherein the heterologous nucleic acid encodes one or more control sequences suitable for directing expression of the one or more genes in a monocytic cell.
19. The donor monocytic cell line of claim 18, wherein the one or more control sequences comprises a promoter.
20. The donor monocytic cell line of claim 15, wherein the cells comprise a vector encoding one or more genes selected from CA2, CLCN7, CTSK, CSF1R, IKBKG, ITGB3, OSTM1, PLEKHM1, TCIRG1, TNFRSF11A, and TNFSF11.
21. The donor monocytic cell line of claim 20, wherein the vector is a mammalian expression vector, a lentiviral vector, or transposon vector.
US17/058,830 2018-06-01 2019-05-31 Methods and compositions for the treatment of osteopetrosis Pending US20210205360A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/058,830 US20210205360A1 (en) 2018-06-01 2019-05-31 Methods and compositions for the treatment of osteopetrosis

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201862679553P 2018-06-01 2018-06-01
PCT/US2019/034984 WO2019232426A1 (en) 2018-06-01 2019-05-31 Methods and compositions for the treatment of osteopetrosis
US17/058,830 US20210205360A1 (en) 2018-06-01 2019-05-31 Methods and compositions for the treatment of osteopetrosis

Publications (1)

Publication Number Publication Date
US20210205360A1 true US20210205360A1 (en) 2021-07-08

Family

ID=68698991

Family Applications (1)

Application Number Title Priority Date Filing Date
US17/058,830 Pending US20210205360A1 (en) 2018-06-01 2019-05-31 Methods and compositions for the treatment of osteopetrosis

Country Status (3)

Country Link
US (1) US20210205360A1 (en)
EP (1) EP3801589A4 (en)
WO (1) WO2019232426A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117551619A (en) * 2024-01-10 2024-02-13 广东工业大学 THP-1 cell with high osteoclast differentiation capability, osteoclast, preparation and application

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140127268A1 (en) * 2007-01-11 2014-05-08 Inserm (Institut National De La Sante Et De La Recherche Medicale) Method for Expanding Monocytes
US20150353890A1 (en) * 2013-01-08 2015-12-10 Fred Hutchinson Cancer Research Center Compositions and methods for expansion of embryonic hematopoietic stem cells

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9050315B2 (en) 2010-04-12 2015-06-09 The Trustees Of The University Of Pennsylvania In vivo and ex vivo expansion of hematopoietic stem cells with a targeted combination of clinically tested, FDA approved drugs
JP2015523058A (en) * 2012-05-10 2015-08-13 バイオマトセル アクチエボラグBiomatcell AB Osteogenic differentiation of mesenchymal stem cells
US10881611B2 (en) * 2015-06-16 2021-01-05 Fondazione Città Della Speranza—Onlus Extracellular vesicles derived from osteoblastic lineage cells for therapeutic and diagnostic use

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20140127268A1 (en) * 2007-01-11 2014-05-08 Inserm (Institut National De La Sante Et De La Recherche Medicale) Method for Expanding Monocytes
US20150353890A1 (en) * 2013-01-08 2015-12-10 Fred Hutchinson Cancer Research Center Compositions and methods for expansion of embryonic hematopoietic stem cells

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Kodama et al. (1991, J. Exp. Med., Vol. 173, pgs. 269-272) (Year: 1991) *
Motyckova et al. (2001, PNAS, Vol. 98(10), pgs. 5798-5803). (Year: 2001) *
Wu et al. (2017, J. Clin. Endocrinol. Metab., Vol. 102(9), pgs. 3111-3123) (Year: 2017) *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117551619A (en) * 2024-01-10 2024-02-13 广东工业大学 THP-1 cell with high osteoclast differentiation capability, osteoclast, preparation and application

Also Published As

Publication number Publication date
EP3801589A4 (en) 2022-03-02
WO2019232426A1 (en) 2019-12-05
EP3801589A1 (en) 2021-04-14

Similar Documents

Publication Publication Date Title
Jacome-Galarza et al. Developmental origin, functional maintenance and genetic rescue of osteoclasts
Ke et al. Embryogenesis and adult life in the absence of intrinsic apoptosis effectors BAX, BAK, and BOK
Pridans et al. Pleiotropic impacts of macrophage and microglial deficiency on development in rats with targeted mutation of the Csf1r locus
Lunney et al. Importance of the pig as a human biomedical model
Khalil et al. Fibroblast-specific TGF-β–Smad2/3 signaling underlies cardiac fibrosis
Chitu et al. Regulation of embryonic and postnatal development by the CSF-1 receptor
Yin et al. Otud7a knockout mice recapitulate many neurological features of 15q13. 3 microdeletion syndrome
Navankasattusas et al. The netrin receptor UNC5B promotes angiogenesis in specific vascular beds
Lever et al. Parabiosis reveals leukocyte dynamics in the kidney
Xue et al. Gene editing in a Myo6 semi-dominant mouse model rescues auditory function
Tucker Dieseases of the Wistar Rat
CN110100788A (en) Methods and applications based on gene manipulation strategy building disease model
Novak et al. Osteoclasts derive predominantly from bone marrow–resident CX3CR1+ precursor cells in homeostasis, whereas circulating CX3CR1+ cells contribute to osteoclast development during fracture repair
Yu et al. Nuclear factor of activated T cells 2 is required for osteoclast differentiation and function in vitro but not in vivo
Stein et al. Massive osteopetrosis caused by non-functional osteoclasts in R51Q SNX10 mutant mice
US20210205360A1 (en) Methods and compositions for the treatment of osteopetrosis
Liu et al. Molecular and cellular mechanisms of the first social relationship: A conserved role of 5-HT from mice to monkeys, upstream of oxytocin
Ferrer-Ferrer et al. Mice deficient in synaptic protease neurotrypsin show impaired spaced long-term potentiation and blunted learning-induced modulation of dendritic spines
Fujino et al. Generation and mutational analysis of a transgenic murine model of the human MAF mutation
US20190142868A1 (en) Methods of treating diseases associated with ilc3 cells
Maes et al. Mitochondrial network adaptations of microglia reveal sex-specific stress response after injury and UCP2 knockout
WO2011035239A1 (en) Kcnq1 and kcne2 in thyroid disease
Barnea et al. Massive osteopetrosis caused by giant, non-functional osteoclasts in R51Q SNX10 mutant mice
Barnea et al. R51Q SNX10 induces osteopetrosis by promoting uncontrolled fusion of monocytes to form giant, non-functional osteoclasts
Gullard et al. Reduced Dentin Matrix Protein Expression in Camurati‐Engelmann Disease Transgenic Mouse Model

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION UNDERGOING PREEXAM PROCESSING

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

AS Assignment

Owner name: NATIONAL INSTITUTES OF HEALTH (NIH), U.S. DEPT. OF HEALTH AND HUMAN SERVICES (DHHS), U.S. GOVERNMENT, MARYLAND

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:SLOAN-KETTERING INST CAN RESEARCH;REEL/FRAME:065365/0471

Effective date: 20211118

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED